Language selection

Search

Patent 3181350 Summary

Third-party information liability

Some of the information on this Web page has been provided by external sources. The Government of Canada is not responsible for the accuracy, reliability or currency of the information supplied by external sources. Users wishing to rely upon this information should consult directly with the source of the information. Content provided by external sources is not subject to official languages, privacy and accessibility requirements.

Claims and Abstract availability

Any discrepancies in the text and image of the Claims and Abstract are due to differing posting times. Text of the Claims and Abstract are posted:

  • At the time the application is open to public inspection;
  • At the time of issue of the patent (grant).
(12) Patent Application: (11) CA 3181350
(54) English Title: TETRAZOLE DERIVATIVES AS TRPA1 INHIBITORS
(54) French Title: DERIVES DE TETRAZOLE UTILES EN TANT QU'INHIBITEURS DE TRPA1
Status: Application Compliant
Bibliographic Data
(51) International Patent Classification (IPC):
  • A61K 31/506 (2006.01)
  • A61K 31/5377 (2006.01)
  • A61P 11/00 (2006.01)
(72) Inventors :
  • BINDER, FLORIAN PAUL CHRISTIAN (Germany)
  • FLECK, MARTIN THOMAS (Germany)
  • WILLWACHER, JENS (Germany)
(73) Owners :
  • BOEHRINGER INGELHEIM INTERNATIONAL GMBH
(71) Applicants :
  • BOEHRINGER INGELHEIM INTERNATIONAL GMBH (Germany)
(74) Agent: SMART & BIGGAR LP
(74) Associate agent:
(45) Issued:
(86) PCT Filing Date: 2021-06-25
(87) Open to Public Inspection: 2022-01-06
Availability of licence: N/A
Dedicated to the Public: N/A
(25) Language of filing: English

Patent Cooperation Treaty (PCT): Yes
(86) PCT Filing Number: PCT/EP2021/067470
(87) International Publication Number: EP2021067470
(85) National Entry: 2022-10-26

(30) Application Priority Data:
Application No. Country/Territory Date
20182988.4 (European Patent Office (EPO)) 2020-06-29

Abstracts

English Abstract

The present disclosure provides certain tetrazole derivatives that are inhibitors of transient receptor potential ankyrin 1 (TRPA1), and are therefore useful for the treatment of diseases treatable by inhibition of TRPA1. Also provided are pharmaceutical compositions containing the same, and processes for preparing said compounds.


French Abstract

La présente invention concerne certains dérivés de tétrazole qui sont des inhibiteurs de l'ankyrine à potentiel de récepteur transitoire 1 (TRPA1) et sont par conséquent utiles pour le traitement de maladies pouvant être traitées par inhibition de TRPA1. L'invention concerne également des compositions pharmaceutiques les contenant et des procédés de préparation desdits composés.

Claims

Note: Claims are shown in the official language in which they were submitted.


CLAIMS
1. A compound according to formula (I)
<IMG>
wherein
A is selected from the group consisting of phenyl, thiophenyl, benzothiophenyl
or benzo-
furanyl, unsubstituted or substituted with one, two or three members of the
group R3 con-
sisting of halogen, C14-alkyl, C14-fluoroalkyl, C3_4-cycloalkyl, C3_4-
cyclofluoroalkyl, 0-
C14-alkyl, 0-cyclopropyl and NC-;
or
A is selected from the group consisting of
<IMG>
is selected from the group consisting of Cl4-alkyl, Cl4-fluoroa1ky1, C3-6-
cycloalkyl, R4-
(H2C).- and R5-(H2qn-;
-98-

wherein
m is 1 or 2;
n is 2;
R4 is C3_6-cycloalkyl;
le is -0-C14-alkyl or -0-Ci4-fluoroalkyl ;
R2 is selected from the group consisting of H, Ci4-alkyl, C3_6-cycloalkyl,
C3_6-cyclofluoro-
alkyl, HO-Ci4-alkyl-, Ci4-fluoroalkyl, R6-(H2C)p-, R7-(H2C)q-, R6-(H(R8)C)p-
and R7-
(H(R9)C)q-;
wherein
p is 1 or 2;
q is 2;
R6 is selected from the group consisting of HO-Ch2-alkyl-, C3_6-cycloalkyl, C-
morpholinyl,
C-imidazolyl and C-pyrazolyl;
wherein said C-pyrazolyl, C-imidazolyl and C-morpholinyl is unsubstituted or
substituted
with Ci4-alkyl or Ci4-fluoroalkyl;
R7 is selected from the group consisting of C14-alky1-0-, C14-fluoroalky1-0-,
Ci4-alkyl-
S(0)2-, N-morpholinyl, N-imidazolyl and N-pyrazolyl;
wherein said N-pyrazolyl, N-imidazolyl, N-morpholinyl is unsubstituted or
substituted
with Ci4-alkyl or Ci4-fluoroalkyl;
le and R9 are independently selected from H or Ci4-alkyl.
2. The compound of formula (I) according to claim 1, wherein A is selected
from the group
consisting of phenyl, thiophenyl, benzothiophenyl or benzofuranyl,
unsubstituted or substi-
tuted with one or two members of the group R3 consisting of Cl, F, Br, H3C,
H3C-0- and
NC-;
or
A is
<IMG>
-99-

3. The compound of formula (I) according to claim 1, wherein A is selected
from the group
consisting of
<IMG>
unsubstituted or substituted with one or two members of the group R3
consisting of Cl, F,
Br, H3C, H3C-0- and NC-,
or
A is
<IMG>
4. The compound of formula (I) according to any of claims 1 to 3, wherein R1
is selected
from the group consisting of C14-alkyl, C3_6-cycloalkyl, R4-(H2C)m- and R5-
(H2C)n-;
wherein
m is 1;
n is 2;
R4 is C3-6-cycloalkyl; and
R5 is -0-C14-alkyl.
-100-

5. The compound of formula (I) according to any of claims 1 to 3, wherein R1
is selected
from the group consisting of C14-alkyl, C34-cycloalkyl, R4-(H2C)m- and R5-
(H2C)n-;
wherein
m is 1;
n is 2;
R4 is C34-cycloalkyl; and
R5 is -0-C14-alkyl.
6. The compound of formula (I) according to any of claims 1 to 5, wherein R2
is selected
from the group consisting of H, Ci4-alkyl, C3_6-cycloalkyl, HO-Ci_4-alkyl-,
Ci_4-fluoroal-
kyl, R6-(H2C)p- and R7-(H2C)q-;
wherein
p is 1;
q is 2;
R6 is selected from the group consisting of C3_6-cycloalkyl, C-morpholinyl, C-
imidazolyl
and C-pyrazolyl;
wherein said C-pyrazolyl, C-imidazolyl and C-morpholinyl is unsubstituted or
substituted
with C 1-4-al kyl.
R7 is selected from the group consisting of -0-C14-alkyl, -0-C14-fluoroalkyl,
C14-alkyl-
S(0)2-, N-morpholinyl, N-imidazolyl and N-pyrazolyl;
wherein said N-pyrazolyl, N-imidazolyl, N-morpholinyl is unsubstituted or
substituted
with C 1-4-al kyl.
7. The compound of formula (I) according to any of claims 1 to 5, wherein R2
is selected
from the group consisting of H, Ci4-alkyl, C3_6-cycloalkyl, HO-Ci_4-alkyl-,
Ci_2-fluoroal-
kyl, R6-(H2C)p- and R7-(H2C)q-;
wherein
p is 1;
q is 2;
R6 is selected from the group consisting of C3_6-cycloalkyl, C-morpholinyl, C-
imidazolyl
and C-pyrazolyl;
-101-

wherein said C-pyrazolyl, C-imidazolyl and C-morpholinyl is unsubstituted or
substituted
with H3C;
R7 is selected from the group consisting of H3C-0, -0-fluoromethyl, H3C-S(0)2-
, N-mor-
pholinyl, N-imidazolyl and N-pyrazolyl;
wherein said N-pyrazolyl, N-imidazolyl, N-morpholinyl is unsubstituted or
substituted
with H3C.
8. The compound of formula (I) according to any of claims 1 to 5, wherein R2
is selected
from the group consisting of H, Ci4-alkyl, C3_6-cycloalkyl, HO-Ci4-alkyl-,
Ci_2-fluoroal-
kyl, R6-(H2C)p- and R7-(H2C)q-;
wherein
p is 1;
q is 2;
R6 is selected from the group consisting of C3_6-cycloalkyl,
<IMG>
R7 is selected from the group consisting of H3C-0, -0-fluoromethyl, H3C-S(0)2-
;
<IMG>
-102-

9. The compound of formula (I) according to any of claims 1 to 3, selected
from the group
consisting of
<IMG>
-103-

<IMG>
-104-

<IMG>
-105-

<IMG>
-106-

<IMG>
10. The compound of formula (I) according to claim 1, selected from the group
consisting
of
-107-

<IMG>
-108-

<IMG>
-109-

<IMG>
-110-

<IMG>
-111-

<IMG>
-112-

<IMG>
-113-

<IMG>
,
-114-

<IMG>
-115-

<IMG>
-116-

<IMG>
-117-

<IMG>
-118-

<IMG>
11. A salt, particularly a pharmaceutically acceptable salt, of a compound
according to any
one of claims 1 to 10.
12. A pharmaceutical composition comprising at least one compound of formula I
accord-
ing to any one of claims 1 to 10 or a pharmaceutically acceptable salt thereof
and one or
more pharmaceutically acceptable excipients.
13. The compound of formula (I) according to one or more of claims 1 to 10, or
a pharma-
ceutically acceptable salt thereof, for use as a medicament.
14. The compound according to any of claims 1 to 10, or a pharmaceutically
acceptable
salt thereof, for the treatment or prevention of inflammatory airway diseases
or fibrotic dis-
eases or cough.
15. The compound according to any of claims 1 to 10, or a pharmaceutically
acceptable
salt thereof, for the treatment or prevention of idiopathic lung disease (IPF)
or cough.
-119-

Description

Note: Descriptions are shown in the official language in which they were submitted.


CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Tetrazole Derivatives as TRPA1 Inhibitors
FIELD OF THE INVENTION
The present disclosure provides certain tetrazole derivatives that are
inhibitors of transient
receptor potential ankyrin 1 (TRPA1), and are therefore useful for the
treatment of diseases
treatable by inhibition of TRPA1. Also provided are pharmaceutical
compositions contain-
ing the same, and processes for preparing said compounds.
BACKGROUND INFORMATION
Transient receptor potential channels (TRP channels) are a group of voltage-
gated ion
channels located mostly on the plasma membrane of numerous mammalian cell
types.
is There are approximately 30 structurally related TRP channels sorted into
groups: TRPA,
TRPC, TRPM, TRPML, TRPN, TRPP and TRPV. Transient receptor potential cation
channel, subfamily A, member 1 (TRPA1), also known as transient receptor
potential
ankyrin 1, is the only member of the TRPA gene subfamily. Structurally, TRPA
channels
are characterized by multiple N-terminal ankyrin repeats (-14 in the N-
terminus of human
zo TRPA1) that gives rise to the "A" for ankyrin designation (Monte11,
2005).
TRPA1 is highly expressed in the plasma membrane of sensory neurons in the
dorsal root
and nodose ganglia that serve both skin and lung, as well as in small
intestine, colon, pan-
creas, skeletal muscle, heart, brain, bladder and lymphocytes
(https://www.proteinat-
las.org/) as well as in human lung fibroblasts.
25 TRPA1 is best known as a sensor for environmental irritants giving rise
to somatosensory
modalities such as pain, cold and itch. TRPA1 is activated by a number of
reactive, elec-
trophilic stimuli (e.g. allyl isothiocyanate, reactive oxygen species), as
well as non-reactive
compounds (e.g. icilin), implicated in cough associated with asthma, chronic
pulmonary
obstructive disease (COPD), idiopathic pulmonary fibrosis (IPF) or post-viral
cough or for
30 chronic idiopathic cough as well as cough in sensitive patients. (Song
and Chang, 2015;
Grace and Belvisi, 2011). TRPA1 inhibitors are useful in the treatment of IPF
in which
cough is highly prevalent because of the link between cough and lung injury,
based on
studies showing cough-induced elevation of TGF-I3 (Xie et al., 2009; Froese et
al., 2016;
-1-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Tschumperlin et al., 2003; Yamamoto et al., 2002; Ahamed et al., 2008). TRPA1
antago-
nists inhibit calcium signaling triggered by cough triggers such as cigarette
smoke extract
(CSE) oxidative stress, inflammatory mediator release and downregulated
antioxidant gene
expression (Lin et al., 2015; Wang et al., 2019). TRPA1 antagonists are
effective in studies
of atopic dermatitis (Oh et al., 2013; Wilson et al., 2013), contact
dermatitis (Liu et al.,
2013), psoriasis-associated itch (Wilson et al., 2013) and IL-31-dependent
itch (Cevikbas
et al., 2014). A human TRPA1 gain-of-function has been associated with
familial episodic
pain syndrome (Kremeyer et al., 2010). A TRPA1 antagonist was effective in a
behavioral
model of migraine-related allodynia (Edelmayer et al., 2012). TRPA1 is
selectively in-
creased in trigeminal ganglia innervating injured teeth when compared to TRPA1
expres-
sion in trigeminal ganglia innervating healthy teeth (Haas et al., 2011).
Several anaesthet-
ics are known to be TRPA1 agonists, including isoflurane (Matta et al., 2008)
providing
rationale for TRPA1 inhibitors for the relief of post-surgical pain. TRPA1
knockout mice
and wild type mice treated with a TRPA1 antagonist showed anxiolytic- and
antidepres-
is sant-like phenotypes (de Moura et al., 2014). TRPA1 inhibitors are
expected to have bene-
fit in the treatment of diabetic neuropathy based on studies showing a
mechanistic link of
inverse regulation between AlVIPK and TRPA1 (Hiyama et al., 2018; Koivisto and
Pertovaara, 2013; Wang et al., 2018). TRPA1 knockout mice exhibit smaller
myocardial
infarct sizes compared to wild type mice (Conklin et al., 2019). TRPA1
knockout and
zo pharmacological intervention inhibited TNBS-induced colitis in mice
(Engel et al., 2011).
In a mouse brain ischaemia model, TRPA1 knock-out and TRPA1 antagonists reduce
mye-
lin damage (Hamilton et al., 2016). Urate crystals and joint inflammation are
reduced in
TRPA1 knockout mice in a monosodium urate mouse model of gout (Moilanen et
al.,
2015). TRPA1 deletion in rats ameliorated joint inflammation and hyperalgesia
in a rat
25 model of acute gout flares (Trevisan et al., 2014). Activation of TRPA1
elicits an inflam-
matory response in osteoarthritic chondrocytes (Nummenmaa et al., 2016). TRPA1
inhibi-
tion and genetic deletion reduces inflammatory mediators in osteoarthritic
mouse chondro-
cytes and murine cartilage (Nummenmaa et al., 2016). Finally, TRPA1 knockout
mice ex-
hibited improvements in weight bearing on the osteoarthritic limb in an MIA-
evoked knee
30 swelling model (Horvath et al., 2016). TRPA1 is differentially expressed
in the bladder ep-
ithelium of rats (Du et al., 2007) and of patients with bladder outlet
obstruction (Du et al.,
2008). TRPA1 receptor modulation attenuates bladder overactivity in a rat
model of spinal
-2-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
cord injury (Andrade et al., 2011) and intrathecal administration of TRPA1
antagonists at-
tenuate cyclophosphamide-induced cystitis in rats with hyper-reflexia
micturition (Chen et
al., 2016).
.. It is therefore desirable to provide potent TRPA1 inhibitors.
TRPA1 inhibitors of various structural classes are reviewed in S. Skerratt,
Progress in Me-
dicinal Chemistry, 2017, Volume 56, 81-115 and in D. Preti, G. Saponaro, A.
Szallasi,
Pharm. Pat. Anal. (2015) 4 (2), 75-94.
W02017/060488 discloses compounds that are antagonists of TRPA1, having the
general-
ized structural formula
0
F py,
y,41=r;
le e t y7 ¨Q
."`""*G8
G2
The TRPA1 activity of Examples 28 and 29 bearing a tetrazolyl ring therein is
not dis-
is closed.
L. Schenkel, et al.,J. Med. Chem. 2016, 59, 2794-2809 discloses quinazolinone-
based
TRPA1 antagonists including compounds of the generalized structural formula
N N
0¨N
CI
0
zo of which compound 31, wherein R is OH, is disclosed as having an
antagonistic TRPA1
activity of ICso 58 nM in a FLIPR assay and having an intrinsic clearance in
human liver
microsomes of <14 L/min/kg.
DETAILED DESCRIPTION OF THE INVENTION
-3-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
The present invention discloses novel tetrazole derivatives that are
inhibitors of transient
receptor potential ankyrin 1 (TRPA1), possessing appropriate pharmacological
and phar-
macokinetic properties enabling their use as medicaments for the treatment of
conditions
and/or diseases treatable by inhibition of TRPA1.
The compounds of the present invention may provide several advantages, such as
en-
hanced potency, high metabolic and/or chemical stability, high selectivity,
safety and toler-
ability, enhanced solubility, enhanced permeability, desirable plasma protein
binding, en-
hanced bioavailability, suitable pharmacokinetic profiles, and the possibility
to form stable
salts.
The compounds of the invention
The present invention provides novel tetrazole derivatives that are
surprisingly potent in-
hibitors of TRPA1 (Assay A), further characterised by
- improved stability in human liver microsomes (Assay B)
- improved stability in human hepatocytes (Assay C).
Compounds of the present invention differ structurally from examples 28 and 29
in
W02017/060488 in that they contain a monocyclic dioxodihydropyrimidine core
with N-
substituents, amido substituents as well as substituents adjacent to a
secondary aliphatic al-
cohol. Compounds of the present invention additionally differ structurally
from example
zo .. 31 in L. Schenkel, et at., J. Med. Chem. 2016, 59, 2794-2809, in that
they bear a tetrazolyl
ring. These structural differences unexpectedly lead to a favourable
combination of (i) in-
hibition of TRPA1, (ii) stability in human liver microsomes and (iii)
stability in human
hepatocytes.
Compounds of the invention are thus superior to those disclosed in the prior
art in terms of
the combination of the following parameters:
-potency as inhibitors of TRPA1
-stability in human liver microsomes
-stability in human hepatocytes
Stability in human liver microsomes refers to the susceptibility of compounds
to biotrans-
formation in the context of selecting and/or designing drugs with favorable
pharmacoki-
netic properties as a first screening step. The primary site of metabolism for
many drugs is
-4-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
the liver. Human liver microsomes contain the cytochrome P450s (CYPs), and
thus repre-
sent a model system for studying phase I drug metabolism in vitro. Enhanced
stability in
human liver microsomes is associated with several advantages, including
increased bioa-
vailability and adequate half-life, which can enable lower and less frequent
dosing of pa-
s tients. Thus, enhanced stability in human liver microsomes is a favorable
characteristic for
compounds that are to be used for drugs. Therefore, compounds of the present
invention in
addition to being able to inhibit TRPA1 are expected to have a favorable in
vivo clearance
and thus the desired duration of action in humans.
Stability in human hepatocytes refers to the susceptibility of compounds to
biotransfor-
io mation in the context of selecting and/or designing drugs with favorable
pharmacokinetic
properties. The primary site of metabolism for many drugs is the liver. Human
hepatocytes
contain the cytochrome P450s (CYPs) and other drug metabolizing enzymes, and
thus rep-
resent a model system for studying drug metabolism in vitro. (Importantly, in
contrast to
liver microsomes assay, the hepatocytes assay covers also phase II
biotransformations as
is well as liver-specific transporter-mediated processes, and therefore
represents a more com-
plete system for drug metabolism studies). Enhanced stability in human
hepatocytes is as-
sociated with several advantages, including increased bioavailability and
adequate half-
life, which can enable lower and less frequent dosing of patients. Thus,
enhanced stability
in human hepatocytes is a favorable characteristic for compounds that are to
be used for
zo drugs.
The present invention provides novel compounds according to formula (I)
0
'N
A---
N--N
HO
H NO
R2
(I)
wherein
-5-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
A is selected from the group consisting of phenyl, thiophenyl, benzothiophenyl
or benzo-
furanyl, unsubstituted or substituted with one, two or three members of the
group R3 con-
sisting of halogen, C14-alkyl, C14-fluoroalkyl, C3_4-cycloalkyl, C3_4-
cyclofluoroalkyl, -0-
C14-alkyl, -0-cyclopropyl and NC-;
or
A is selected from the group consisting of
* = 0
0
* = 0
0
and
* 0
0 F
=
R1 is selected from the group consisting of C14-alkyl, C14-fluoroalkyl, C3_6-
cycloalkyl, R4-
(H2C)m- and R5-(H2C)n-;
wherein
m is 1 or 2;
n is 2;
is .. R4 is C3_6-cycloalkyl;
R5 is -0-C14-alkyl or -0-C14-fluoroalkyl;
R2 is selected from the group consisting of H, C14-alkyl, C3_6-cycloalkyl,
C3_6-cyclofluoro-
alkyl, HO-C14-alkyl-, C14-fluoroalkyl, R6-(H2C)p-, R7-(H2C)q-, R6-(H(R8)C)-
and R7-
(H(R9)C)q-;
zo wherein
pis 1 or 2;
q is 2;
R6 is selected from the group consisting of HO-C1_2-alkyl-, C3_6-cycloalkyl, C-
morpholinyl,
C-imidazolyl and C-pyrazolyl;
-6-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
wherein said C-pyrazolyl, C-imidazolyl and C-morpholinyl is unsubstituted or
substituted
with Ci4-alkyl or Ci4-fluoroalkyl;
R7 is selected from the group consisting of -0-C14-alkyl, -0-C14-fluoroalkyl,
C14-alkyl-
S(0)2-, N-morpholinyl, N-imidazolyl and N-pyrazolyl;
.. wherein said N-pyrazolyl, N-imidazolyl, N-morpholinyl is unsubstituted or
substituted
with Ci4-alkyl or Ci4-fluoroalkyl;
R8 and le are independently selected from H or C14-alkyl.
Another embodiment of the present invention relates to a compound of formula
(I),
io wherein
A is selected from the group consisting of phenyl, thiophenyl, benzothiophenyl
or benzo-
furanyl, unsubstituted or substituted with one or two members of the group R3
consisting
of halogen, C14-alkyl, -0-C14-alkyl and NC-;
or
A is
* 0
0
=
zo R1 is selected from the group consisting of C14-alkyl, C3_6-cycloalkyl,
R4-(H2C)m- and R5-
(H2C)n-;
wherein
m is 1 or 2;
n is 2;
.. R4 is C3_6-cycloalkyl;
R5 is -0-C14-alkyl;
R2 is selected from the group consisting of H, C14-alkyl, C3_6-cycloalkyl, HO-
C14-alkyl-,
Ci4-fluoroalkyl, R6-(H2C)p- and le-(H2C)q-;
wherein
pis 1 or 2;
-7-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
q is 2;
R6 is selected from the group consisting of C3_6-cycloalkyl, C-morpholinyl, C-
imidazolyl
and C-pyrazolyl;
wherein said C-pyrazolyl, C-imidazolyl and C-morpholinyl is unsubstituted or
substituted
with C14-alkyl;
R7 is selected from the group consisting of ¨0-C14-alkyl, -0-C14-fluoroalkyl,
C14-alkyl-
S(0)2-, N-morpholinyl, N-imidazolyl and N-pyrazolyl;
wherein said N-pyrazolyl, N-imidazolyl, N-morpholinyl is unsubstituted or
substituted
io with C14-alkyl.
Another embodiment of the present invention relates to a compound of formula
(I)
wherein
A is selected from the group consisting of phenyl, thiophenyl, benzothiophenyl
or benzo-
is furanyl, unsubstituted or substituted with one or two members of the
group R3 consisting
of Cl, F, Br, H3C, H3C-0- and NC-;
or
20 A is
* 0
0
=
and substituents R1 and R2 are defined as in the preceding embodiment.
Another embodiment of the present invention relates to a compound of formula
(I)
25 wherein
A is selected from the group consisting of
* =
-8-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
*
*
* 1011
0
and
0
unsubstituted or substituted with one or two members of the group R3
consisting of Cl, F,
Br, H3C, H3C-0- and NC-,
or
A is
* 0
0
and substituents R1 and R2 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
A is selected from the group consisting of
* =
* =
-9-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
* = CI
,
* . Br
,
*-qS
CI
,
*MNS
N N
,
CI
*-eXS
CI
,
* 1 0S
,
* 1 10
,
* 1 .S F
,
* . 0
F
,
*
CI
/ I.0
,
-10-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
*
0
*
0
*
0
and
* 0
0
and substituents R1 and R2 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
R1 is selected from the group consisting of C14-alkyl, C3_6-cycloalkyl, R4-
(H2C).- and R5-
(1-12C)o-;
wherein
m is 1;
n is 2;
R4 is C3_6-cycloalkyl;
R5 is -0-C14-alkyl;
and substituents A and R2 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
zo R1 is selected from the group consisting of C14-alkyl, C34-cycloalkyl,
R4-(H2C).- and R5-
(1-12C)n-;
wherein
m is 1;
-11-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
n is 2;
R4 is C3_4-cycloalkyl;
R5 is -0-C1_4-alkyl;
and substituents A and R2 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
R1 is selected from the group consisting of C1_4-alkyl, C3_4-cycloalkyl, R4-
(H2C).- and R5-
(H2C)n-;
io wherein
m is 1;
n is 2;
R4 is C3_4-cycloalkyl;
R5 is H3C-O-;
is and substituents A and R2 are defined as in any of the preceding
embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
R1 is selected from the group consisting of H3C, H3CH2C, H3C0H2CH2C,
1 6
= 20 and
and substituents A and R2 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
25 is H3C;
and substituents A and R2 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
30 R2 is selected from the group consisting of H, C3_6-cycloalkyl,
R6-(H2C)p- and le-(H2C)q-;
-12-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
wherein
p is 1;
q is 2;
R6 is selected from the group consisting of C3_6-cycloalkyl, C-morpholinyl, C-
imidazolyl
and C-pyrazolyl;
wherein said C-pyrazolyl, C-imidazolyl and C-morpholinyl is unsubstituted or
substituted
with C14-alkyl;
R7 is selected from the group consisting of -0-C14-alkyl, -0-C14-fluoroalkyl,
C14-alkyl-
S(0)2-, N-morpholinyl, N-imidazolyl and N-pyrazolyl;
io wherein said N-pyrazolyl, N-imidazolyl, N-morpholinyl is unsubstituted
or substituted
with C14-alkyl;
and substituents A and Ware defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
is wherein
R2 is selected from the group consisting of H, C14-alkyl, C3_6-cycloalkyl, HO-
C14-alkyl-,
C1_2-fluoroalkyl, R6-(H2C)p- and le-(H2C)q-;
wherein
p is 1;
zo .. q is 2;
R6 is selected from the group consisting of C3_6-cycloalkyl, C-morpholinyl, C-
imidazolyl
and C-pyrazolyl;
wherein said C-pyrazolyl, C-imidazolyl and C-morpholinyl is unsubstituted or
substituted
with H3 C ;
25 R7 is selected from the group consisting of H3C-0-, -0-fluoromethyl, H3C-
S(0)2-, N-mor-
pholinyl, N-imidazolyl and N-pyrazolyl;
wherein said N-pyrazolyl, N-imidazolyl, N-morpholinyl is unsubstituted or
substituted
with H3 C ;
and substituents A and R1 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
-13-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
R2 is selected from the group consisting of H, C1_4-alkyl, C3_6-cycloalkyl, HO-
C1_4-alkyl-,
C12-fluoroalkyl, R6-(H2C)p- and R7-(H2C)q-;
wherein
p is 1;
q is 2;
R6 is selected from the group consisting of C3_6-cycloalkyl,
0
and
(IN
io R7 is selected from the group consisting of H3C-0, -0-fluoromethyl, H3C-
S(0)2-,
C. N
N *
c*.N
and
0
cN
and substituents A and Ware defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
wherein
R2 is selected from the group consisting H, H3C, H3CH2C, H3C0H2CH2C, F2HCH2C,
F3CH2C, FH2CH2C, H3C(0)2SH2CH2C, F3C0H2CH2C,
-14-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
CNI
N
N
I
N
0
0
C
and
and substituents A and R1 are defined as in any of the preceding embodiments.
Another embodiment of the present invention relates to a compound of formula
(I),
io wherein
R2 is H;
and substituents A and R1 are defined as in any of the preceding embodiments.
Preferred is a compound of formula (I), selected from the group consisting of
-15-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
NNVYNA
/
N-7-----N
0 HO
ONH2
,
0
NNI.NrNrA
N=N/ OH
0
ONH2
,
1 0
NNyNNirA
OH
0
ONH2
,
0
N N rNLN/N.i/A
N=--N OH
0
ON H2
,
0
N.,% ,...-=\,
N NINrNINN
N-I :(H A
0
ON H2
,
-16-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0
..õ.õ.0,.,,...õ.õ..--....,le,=N.,,N.....õ....,yN.N,..õ.yA
N=--I OH
C)
ON H2
,
0
IN="-----N
0 HO
HN'O
..
0
/
,
0
NNIVrN\N"---,A
N.---4
o/
HO
NO
F __________ K H
,
0
NN7rN\NIMA
i
IN -=----N
C 0 HO
\ NN0
H
,
-17-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
A
HO
HNO
\o/
0
NNIVrN\N')A
o/
HO
HN 0
0
NNIVrNi\NIA
0 HO
0
NNIVrNNN-'")____A
o/
HO
ONH
-18-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
N,
A
0 0 HO
I I
*NO
0
0
A
o/
HO
\N/0
0
A
N--N/
c) HO
HN'O
FX0
0
A
o/
HO
x¨NH
-19-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0
NN
INVrN\A
0 HO
0
NNVri\j\NIA
0 HO
FH
and
0
NNVrN\iNA
o/ HO
ONH
and substituent A is defined as in any of the preceding embodiments.
Preferred is the compound according to formula (I) selected from
0
NN7rN\A
o/
HO
ONH2
and substituent A is defined as in any of the preceding embodiments.
-20-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Particularly preferred is the compound according to formula (I) selected from
the group
consisting of
0
XNINIVY1\1\1 = CI
Nz-zzi
0 HO
H2N 0
,
0
NN7rN\N /
/
0 HO
ON H2
,
F
0
S.
N7Nri\IN
/
N=_-_,N OH
0
ON H2
,
-21-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
F
0 -.....,
HC;
0
~ N.,'
0
H2N'O
,
CI
0
i
f\IN.Nr
Nyy(
I
S
o7 N=N OH
C"NH2
,
0 CI
NN7rNI\N /
K 1 /
I NN 0
o HO
ON H2
,
-22-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
. Br
NNVrNNNII
o HO
ON H2
,
0 F
NN7rN\NI /
" /
IN::::=N
o./ 0
HO
ON H2
,
0
N
Xl\I OH
N N /
N_NN2.0
o~
\
H2NO
,
-23-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
N
NN7r \N1---\c
C) HO
ON H2
,
0 0---t
1
0
11\1M-%N\N
o/ Nz:-...i
HO
ON H2
,
0 F
rN,,N 1
/
o/ NN OH
ONH2
,
0
\
'N NNIN
o/ N--N/ OH
ON H2
,
-24-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
ri\i/N j
N N
S-----.N
o~ Nzz-N
HO
oN H2
,
0
Fig
\ 0
o/ N,---_,N/
F
H2NO
,
0 0 CI
N1NNXN
N=N/ OH
C)
ON
H2
,
-25-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
1 0 0 CI
N,
N N.Nr 1\1
NNI OH
0
ON H2
,
0 CI
NNNNN
o/ N--1 OH
oNH2
,
0 0 CI
e''
NI\INNI
o/ NN1 OH
oNH2
,
-26-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0 0 CI
0
NNNr%
N -4 OH
0
0 N H2
,
0
= CI
NN7rN \N
,, /
1 N=_-_-N
0 HO
He%0
0
/
,
0
NNIVYNNI 4Ik CI
/
NN
0 HO
N 0
F_K H
,
-27-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
. CI
NN7rN\N
/
NI-7-----N1
f":-..1 C) HO
...NNO
H
,
0
IN7rNI\N I
/
N:-.-----N
c) HO
HN 0
N
\o/
,
0
NN7rNi\NI = CI
N--z-zil
0 HO
H1"0
C)
=N
,
-28-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
INIVrN\1\1 I
0
HO
N
\
,
0
NNrN\NI = CI
Nz-z--_11
0 HO
ONH
1
,
0
NN7rNI\N I
NI:z__Ni
0 0 HO
I I
S
/IIN 0
0 H
,
-29-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
. CI
NN7rN\N
/
N:------N
c) HO
NC\ NNo
H
,
0
NN7rN\NI I
" /
IN-7-_-__N
0 HO
HN" '0
F 0
FX
F
,
0
NN7r1\1\1\1 = CI
/
Nz--7N
0 HO
N 0
FX H
,
-30-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
0
NN7r\I = CI
/
Nz---N
0 HO
HNO
V)
,
0
= CI
NN7rN\N
,, /
I N=ZN
o
HO
/¨NH'0
F
,
0
. CI
NNVri\IXN,
N:_----N/
(:) HO
ONH
and
-31-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0 0
OH
ON H2
USED TERMS AND DEFINITIONS
Terms not specifically defined herein should be given the meanings that would
be given to
them by one of skill in the art in light of the disclosure and the context. As
used in the
specification, however, unless specified to the contrary, the following terms
have the
meaning indicated and the following conventions are adhered to.
io In the groups, radicals, or moieties defined below, the number of carbon
atoms is often
specified preceding the group, for example, "C1_6-alkyl" means an alkyl group
or radical
having 1 to 6 carbon atoms. In general in groups like HO, H2N, (0)S, (0)2S, NC
(cyano),
HOOC, F3C or the like, the skilled artisan can see the radical attachment
point(s) to the
molecule from the free valences of the group itself. For combined groups
comprising two
is or more subgroups, the last named subgroup is the radical attachment
point, for example,
the substituent "aryl-Ci_3-alkyl" means an aryl group which is bound to a Cii-
alkyl-group,
the latter of which is bound to the core or to the group to which the
substituent is attached.
In case a compound of the present invention is depicted in form of a chemical
name and as
zo a formula in case of any discrepancy the formula shall prevail. An
asterisk may be used in
sub-formulas to indicate the bond which is connected to the core molecule as
defined.
The numeration of the atoms of a substituent starts with the atom that is
closest to the core
or to the group to which the substituent is attached.
For example, the term "3-carboxypropyl-group" represents the following
substituent:
-32-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
1 3
2
0
wherein the carboxy group is attached to the third carbon atom of the propyl
group. The
terms "1-methylpropyl-", "2,2-dimethylpropyl-" or "cyclopropylmethyl-" group
represent
the following groups:
cH3 1 3
2 CH
CH3 3 \-<1
* 1 3
2 H3C CH3
The asterisk may be used in sub-formulas to indicate the bond that is
connected to the core
io molecule as defined.
The term "Ch-alkyl", wherein n is an integer selected from 2, 3, 4 or 5,
either alone or in
combination with another radical denotes an acyclic, saturated, branched or
linear hydro-
carbon radical with 1 to n C atoms. For example the term Cis-alkyl embraces
the radicals
H3 C-, H3 C-CH2-, H3C-CH2-CH2-, H3 C-CH(CH3)-, H3 C-CH2-CH2-CH2-,
H3 C-CH2-CH(CH3)-, H3 C-CH(CH3)-CH2-, H3 C-C(CH3)2-, H3 C-CH2-CH2-CH2-CH2-,
H3 C-CH2-CH2-CH(CH3)-, H3 C-CH2-CH(CH3)-CH2-, H3 C-CH(CH3)-CH2-CH2-,
H3 C-CH2-C(CH3)2-, H3 C-C(CH3)2-CH2-, H3 C-CH(CH3)-CH(CH3)- and
H3 C-CH2-CH(CH2CH3)-.
The term "fluoro" added to an "alkyl", "alkylene" or "cycloalkyl" group
(saturated or un-
saturated) means such a alkyl or cycloalkyl group wherein one or more hydrogen
atoms are
replaced by a fluorine atom. Examples include, but are not limited to: H2FC-,
HF2C- and
F3C-.
The term "C3_,-cycloalkyl", wherein n is an integer from 4 to n, either alone
or in combina-
tion with another radical denotes a cyclic, saturated, unbranched hydrocarbon
radical with
3 to n C atoms. For example the term C3,6-cycloalkyl includes cyclopropyl,
cyclobutyl, cy-
clopentyl and cyclohexyl.
The term halogen generally denotes fluorine, chlorine, bromine and iodine.
The term "phenyl" refers to the radical of the following ring
-33-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
S.
The term "thiophenyl" refers to the radical of the following ring
ys
ii
The term "benzothiophenyl" refers to the radical of the following ring
s
\ .
The term "benzofuranyl" refers to the radical of the following ring
0
\==
The term "tetrazoly1" refers to the radical of the following ring
eNN
,
N=N
The term "dioxodihydropyrimidinecarboxamide" refers to the radical of the
following core
0
.--......
N..õ.. N
N'O
The term "C-morpholinyl" refers to the radical of the following ring
0
( 1
N *
The term "C-imidazoly1" refers to the radical of the following ring
-34-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
("11\1
The term "C-pyrazoly1" refers to the radical of the following ring
*_CNI
N
The term "N-morpholinyl" refers to the radical of the following ring
0
cN *
The term "N-imidazoly1" refers to the radical of the following ring
N
c NI
\*
The term "N-pyrazoly1" refers to the radical of the following ring
CNI
N
\*
The term "substituted" as used herein, means that any one or more hydrogens on
the desig-
nated atom is replaced with a selection from the indicated group, provided
that the desig-
nated atom's normal valence is not exceeded, and that the substitution results
in a stable
compound.
Unless specifically indicated, throughout the specification and the appended
claims, a
is given chemical formula or name shall encompass tautomers and all stereo,
optical and geo-
metrical isomers (e.g. enantiomers, diastereomers, E/Z isomers etc.) and
racemates thereof
as well as mixtures in different proportions of the separate enantiomers,
mixtures of dia-
stereomers, or mixtures of any of the foregoing forms where such isomers and
enantiomers
exist, as well as salts, including pharmaceutically acceptable salts thereof
and solvates
zo thereof such as for instance hydrates including solvates of the free
compounds or solvates
of a salt of the compound.
In general, substantially pure stereoisomers can be obtained according to
synthetic princi-
ples known to a person skilled in the field, e.g. by separation of
corresponding mixtures, by
using stereochemically pure starting materials and/or by stereoselective
synthesis. It is
-35-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
known in the art how to prepare optically active forms, such as by resolution
of racemic
forms or by synthesis, e.g. starting from optically active starting materials
and/or by using
chiral reagents.
Enantiomerically pure compounds of this invention or intermediates may be
prepared via
asymmetric synthesis, for example by preparation and subsequent separation of
appropriate
diastereomeric compounds or intermediates which can be separated by known
methods
(e.g. by chromatographic separation or crystallization) and/or by using chiral
reagents,
such as chiral starting materials, chiral catalysts or chiral auxiliaries.
Further, it is known to the person skilled in the art how to prepare
enantiomerically pure
io compounds from the corresponding racemic mixtures, such as by
chromatographic separa-
tion of the corresponding racemic mixtures on chiral stationary phases; or by
resolution of
a racemic mixture using an appropriate resolving agent, e.g. by means of
diastereomeric
salt formation of the racemic compound with optically active acids or bases,
subsequent
resolution of the salts and release of the desired compound from the salt; or
by derivatiza-
is tion of the corresponding racemic compounds with optically active chiral
auxiliary rea-
gents, subsequent diastereomer separation and removal of the chiral auxiliary
group; or by
kinetic resolution of a racemate (e.g. by enzymatic resolution); by
enantioselective crystal-
lization from a conglomerate of enantiomorphous crystals under suitable
conditions; or by
(fractional) crystallization from a suitable solvent in the presence of an
optically active chi-
n ral auxiliary.
The phrase "pharmaceutically acceptable" is employed herein to refer to those
compounds,
materials, compositions, and/or dosage forms which are, within the scope of
sound medical
judgment, suitable for use without excessive toxicity, irritation, allergic
response, or other
problem or complication, and commensurate with a reasonable benefit/risk
ratio.
25 As used herein, "pharmaceutically acceptable salt" refers to derivatives
of the disclosed
compounds wherein the parent compound forms a salt or a complex with an acid
or a base.
Examples of acids forming a pharmaceutically acceptable salt with a parent
compound
containing a basic moiety include mineral or organic acids such as
benzenesulfonic acid,
benzoic acid, citric acid, ethanesulfonic acid, fumaric acid, gentisic acid,
hydrobromic
30 acid, hydrochloric acid, maleic acid, malic acid, malonic acid, mandelic
acid, methanesul-
fonic acid, 4-methyl-benzenesulfonic acid, phosphoric acid, salicylic acid,
succinic acid,
sulfuric acid and tartaric acid.
Examples for cations and bases forming a pharmaceutically acceptable salt with
a parent
-36-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
compound containing an acidic moiety include Nat, K+, Ca2+, Mg2+, NH4 +, L-
arginine,
2,2'-iminobisethanol, L-lysine, N-methyl-D-glucamine or tris(hydroxymethyl)-
amino-
methane. The pharmaceutically acceptable salts of the present invention can be
synthesized
from the parent compound that contains a basic or acidic moiety by
conventional chemical
methods. Generally, such salts can be prepared by reacting the free acid or
base forms of
these compounds with a sufficient amount of the appropriate base or acid in
water or in an
organic diluent like ether, ethyl acetate, ethanol, isopropanol, or
acetonitrile, or a mixture
thereof.
Salts of other acids than those mentioned above which for example are useful
for purifying
or isolating the compounds of the present invention (e.g. trifluoroacetate
salts,) also com-
prise a part of the present invention.
BIOLOGICAL ASSAYS
is Evaluation of TRPA1 activity
Assay A: TRPA1 assay
The activity of the compounds of the invention may be demonstrated using the
following
in vitro TRPA1 cell assay:
zo Method:
A human HEK293 cell line over-expressing the human TRPA1 ion channel (Perkin
Elmer,
Product No. AX-004-PCL) is used as a test system for compound efficacy and
potency.
Compound activity is determined by measuring the effect of compounds on
intracellular
calcium concentration induced by AITC (Allylisothiocyanat) agonism in a
FLIPRtetra sys-
25 .. tem (Molecular Devices).
Cell culture:
The cells are obtained as frozen cells in cryo-vials and stored until use at -
150 C.
Cells are grown in culture medium (MEM/EBSS medium with 10% FCS and 0.4mg/ML
30 Geneticin). It is important that density does not exceed 90% confluence.
For sub-culturing
cells are detached from flasks by Versene. At the day before the assay, cells
are detached,
washed twice with medium (MEM/EB SS medium with 10% FCS) and 20000 cells in
20W/well are seeded to Poly D-Lysin biocoated 384-well plates (black, clear
bottom,
Cat.356697) from Corning. Plates are incubated for 24 hours at 37 C/5% CO2
before use
35 in the assay.
-37-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Compound preparation
The test compounds are dissolved in 100 % DMSO at a concentration of 10 mM and
in a
first step diluted in DMSO to a concentration of 5 mM, followed by serial
dilution steps in
100% DMSO. Dilution factor and number of dilution steps may vary according to
needs.
Typically 8 different concentrations by 1:5 dilutions are prepared, further
intermediate di-
lutions (1:20) of the substances are carried out with HBSS/HEPES buffer
(1xHEPES,Cat.14065 from Gibco, 20mM HEPES, Cat. 83264 from SIGMA, 0.1% BSA
Cat.11926 from Invitrogen, pH 7.4
io FLIPR assay:
At the assay day cells are washed 3x with assay puffer, 20
buffer remaining in the wells
after washing. 10 Ca6 kit (Cat.R8191 MolecularDevices) loading buffer in
HBSS/HEPES is added to the cells and the plates are incubated with lid for 120
minutes at
370/5% CO2. 10 tL of compound or controls in HBSS/HEPES buffer/5% DMSO from
the
is intermediate dilution plate are carefully added to the wells.
Luminescence (indicating the
calcium influx or release) is read on the FLIPRtetra device for 10 minutes to
monitor the
compound induced effects (e.g. agonism). Finally 10 tL of the agonist AITC
50[tM dis-
solved in HBSS/HEPES buffer/0.05% DMSO (final concentration 10 l.M) is added
to the
wells followed by an additional read on the FLIPRtetra device for 10 minutes.
The area un-
2.0 .. der the signal curve (AUC) after AITC addition is used for IC50 / %
inhibition calculations
Data evaluation and calculation:
Each assay microtiter plate contains wells with vehicle (1% DMSO) controls
instead of
compound as controls for AITC induced luminescence (100 %CTL; high controls)
and
25 wells with vehicle controls without AITC as controls for non-specific
changes in lumines-
cence (0 %CTL; low controls).
The analysis of the data is performed by the calculation of the area under
signal curve of
the individual wells. Based on this values the % value for the measurement of
each sub-
stance concentration is calculated (AUC(sample) - AUC(low))*100/(AUC(high) -
30 AUC(low)) using MegaLab software (in house development). The IC50 values
are calcu-
lated from the % control values using MegaLab software. Calculation: [y=(a-
d)/(1+(x/c)^13)+4 a = low value, d = high value; x = conc M; c=IC50 M; b =
hill; y = %
ctrl
-38-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
Table 1: Biological data for compounds of the invention as obtained in Assay A
Example hTRPA1 ICso
[nM]
1 40
2 18
3 18
4 34
26
6 42
7 56
8 59
9 72
92
11 105
12 110
13 124
14 156
198
16 160
17 213
18 33
19 62
64
21 258
22 23
23 39
24 39
43
26 100
27 140
28 186
-39-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
29 229
30 44
31 68
32 92
33 116
34 297
35 33
Table 2: Biological data for prior art compounds (examples 28 and 29 in
W02017/060488)
as obtained in Assay A.
Example in hTRPA1 ICso
W02017/060488 [nM]
28 366
29 1120
Table 3: Biological data for prior art compounds (example 31 in L. Schenkel,
et at., J.
Med. Chem. 2016, 59, 2794-2809) as obtained in Assay A.
Example in Med. Chem. hTRPA1 ICso
2016, 59, 2794-2809 [nM]
31 52
Evaluation of Microsomal Clearance
Assay B: Microsomal clearance:
io The metabolic degradation of the test compound is assayed at 37 C with
pooled liver mi-
crosomes. The final incubation volume of 100 11.1 per time point contains TRIS
buffer pH
7.6 at RT (0.1 M), magnesium chloride (5 mM), microsomal protein (1 mg/ml) and
the test
compound at a final concentration of 1 04.
Following a short preincubation period at 37 C, the reactions are initiated by
addition of
is .. beta-nicotinamide adenine dinucleotide phosphate, reduced form (NADPH, 1
mM) and ter-
minated by transferring an aliquot into solvent after different time points
(0, 5, 15, 30, 60
min). Additionally, the NADPH-independent degradation is monitored in
incubations with-
out NADPH, terminated at the last time point. The [%] remaining test compound
after
-40-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
NADPH independent incubation is reflected by the parameter c(control)
(metabolic stabil-
ity). The quenched incubations are pelleted by centrifugation (10000 g, 5
min).
An aliquot of the supernatant is assayed by LC-MS/MS for the amount of parent
com-
pound. The half-life (t1/2 INVITRO) is determined by the slope of the
semilogarithmic
plot of the concentration-time profile.
The intrinsic clearance (CL INTRINSIC) is calculated by considering the amount
of pro-
tein in the incubation:
CL INTRINSIC [ 1/min/mg protein] = (Ln 2 / (half-life [min] * protein content
[mg/m1]))
* 1000
CL INTRINSIC INVIVO [ml/min/kg] = (CL INTRINSIC [ L/min/mg protein] x
MPPGL [mg protein/g liver] x liver factor [g/kg bodyweight]) / 1000
is Qh [%] = CL [ml/min/kg] / hepatic blood flow [ml/min/kg])
Hepatocellularity, human: 120x10e6 cells / g liver
Liver factor, human: 25.7 g / kg bodyweight
Blood flow, human: 21 ml/(min x kg)
Table 4: Biological data for compounds of the invention as obtained in Assay B
Example human LM [%Qh]
1 <23
2 <23
3 26
4 <23
5 <23
6 <23
7 <23
8 <23
9 <23
10 <23
-41-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
11 <23
12 <23
13 <23
14 <23
15 <23
16 <23
17 42
18 38
19 <23
20 <23
21 <23
22 <23
23 <23
24 38
25 55
26 52
27 <23
28 <23
29 53
30 28
31 <23
32 52
33 57
34 43
35 <23
Table 5: Biological data for prior art compounds (examples 28 and 29 in
W02017/060488)
as obtained in Assay B.
Example in human LM [%Qh]
W02017/060488
28 62
29 <23
-42-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Table 6: Biological data for prior art compounds (example 31 in L. Schenkel,
et at., J.
Med. Chem. 2016, 59, 2794-2809) as obtained in Assay B.
Example in Med. Chem. human LM [%Qh]
2016, 59, 2794-2809
31 <23
Evaluation of Hepatocyte Clearance
Assay C: Hepatocyte clearance
The metabolic degradation of the test compound is assayed in a hepatocyte
suspension.
Hepatocytes (cryopreserved) are incubated in Dulbecco's modified eagle medium
(supple-
mented with 3.5[tg glucagon/500mL, 2.5mg insulin/500mL and 3.75mg/500mL
hydrocor-
io tison) containing 5% or 50% species serum.
Following a 30 min preincubation in an incubator (37 C, 10% CO2) 5 11.1 of
test compound
solution (80 [tM; from 2mM in DMSO stock solution diluted 1:25 with medium)
are added
into 39511.1 hepatocyte suspension (cell density in the range 0.25-5 Mio
cells/mL depend-
ing on the species, typically 1 Mio cells/mL; final concentration of test
compound l[tM,
is final DMSO concentration 0.05%).
The cells are incubated for six hours (incubator, orbital shaker) and samples
(25 1) are
taken at 0, 0.5, 1, 2, 4 and 6 hours. Samples are transferred into
acetonitrile and pelleted by
centrifugation (5 min). The supernatant is transferred to a new 96-deepwell
plate, evapo-
rated under nitrogen and resuspended.
zo Decline of parent compound is analyzed by HPLC-MS/MS
CLint is calculated as follows CL INTRINSIC = Dose / AUC = (CO/CD) / (AUD +
clast/k) x 1000/60. CO: initial concentration in the incubation [On CD: cell
density of vi-
tal cells [10e6ce11s/mL], AUD: area under the data [[tM x h], clast:
concentration of last
data point [04], k: slope of the regression line for parent decline [h-1].
25 The calculated in vitro hepatic intrinsic clearance can be scaled up to
the intrinsic in vivo
hepatic Clearance and used to predict hepatic in vivo blood clearance (CL) by
the use of a
liver model (well stirred model).
CL INTRINSIC INVIVO [ml/min/kg] = (CL INTRINSIC [ L/min/10e6cells] x hepato-
30 cellularity [10e6 cells/g liver] x liver factor [g/kg bodyweight]) /
1000
-43-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
CL [ml/min/kg] = CL INTRINSIC INVIVO [ml/min/kg] x hepatic blood flow
[ml/min/kg] / (CL INTRINSIC INVIVO [ml/min/kg] + hepatic blood flow
[ml/min/kg])
Qh [%] = CL [ml/min/kg] / hepatic blood flow [ml/min/kg])
Hepatocellularity, human: 120x10e6 cells / g liver
Liver factor, human: 25.7 g / kg bodyweight
Blood flow, human: 21 ml/(min x kg)
Table 7: Biological data for compounds of the invention as obtained in Assay C
Example human Hepatocytes
[%Qh]
1 <4
2 13
3 27
4 8
5 43
6 15
7 <4
8 6
9 7
10 15
11 <4
12 16
13 <4
14 4
14
16 <4
17 49
18 39
19 5
-44-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
20 15
21 15
22 25
23 15
24 28
25 42
26 21
27 17
28 <4
29 27
30 9
31 4
32 45
33 28
34 44
35 19
Table 8: Biological data for prior art compounds (examples 28 and 29 in
W02017/060488)
as obtained in Assay C.
Example in human Hepatocytes
W02017/060488 [%Qh]
28 49
29 22
Table 9: Biological data for prior art compounds (example 31 in L. Schenkel,
et at., J.
Med. Chem. 2016, 59, 2794-2809) as obtained in Assay C.
Example in Med. Chem. human Hepatocytes
2016, 59, 2794-2809 [%Qh]
31 73
Evaluation of permeability
Caco-2 cells (1 - 2 x 105 cells/1 cm2 area) are seeded on filter inserts
(Costar transwell
io polycarbonate or PET filters, 0.411m pore size) and cultured (DMEM) for
10 to 25 days.
-45-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Compounds are dissolved in appropriate solvent (like DMSO, 1 - 20 mM stock
solutions).
Stock solutions are diluted with HTP-4 buffer (128.13 mM NaCl, 5.36 mM KC1, 1
mM
MgSO4, 1.8 mM CaCl2, 4.17 mM NaHCO3, 1.19 mM Na2HPO4 x 7H20, 0.41 mM
NaH2PO4xH20, 15 mM HEPES, 20 mM glucose, 0.25% BSA, pH 7.2) to prepare the
transport solutions (0.1 -300 [tM compound, final DMSO <= 0.5 %). The
transport solu-
tion (TL) is applied to the apical or basolateral donor side for measuring A-B
or B-A per-
meability (3 filter replicates), respectively. Samples are collected at the
start and end of ex-
periment from the donor and at various time intervals for up to 2 hours also
from the re-
ceiver side for concentration measurement by HPLC-MS/MS or scintillation
counting.
io Sampled receiver volumes are replaced with fresh receiver solution.
Evaluation of plasma protein binding
This equilibrium dialysis (ED) technique is used to determine the approximate
in vitro
fractional binding of test compounds to plasma proteins. Dianorm Teflon
dialysis cells
is (micro 0.2) are used. Each cell consists of a donor and an acceptor
chamber, separated by
an ultrathin semipermeable membrane with a 5 kDa molecular weight cutoff Stock
solu-
tions for each test compound are prepared in DMSO at 1 mM and diluted to a
final concen-
tration of 1.0 [i.M. The subsequent dialysis solutions are prepared in pooled
human or rat
plasma (with NaEDTA) from male and female donors. Aliquots of 200 [IL dialysis
buffer
zo (100 mM potassium phosphate, pH 7.4) are dispensed into the buffer
chamber. Aliquots of
200 [EL test compound dialysis solution are dispensed into the plasma
chambers. Incuba-
tion is carried out for 2 hours under rotation at 37 C.
At the end of the dialysis period, the dialysate is transferred into reaction
tubes. The tubes
for the buffer fraction contain 0.2 mL ACN/water (80/20). Aliquots of 25 [IL
of the plasma
25 dialysate are transferred into deep well plates and mixed with 25 [IL
ACN/water (80/20),
25 [EL buffer, 25 [IL calibration solution and 25 [IL Internal Standard
solution. Protein pre-
cipitation is done by adding 200 [IL ACN. Aliquots of 50 [IL of the buffer
dialysate are
transferred into deep well plates and mixed with 25 [IL blank plasma, 25 [IL
Internal Standard solution and 200 [IL ACN. Samples are measured on HPLC-MS/MS-
30 Systems and evaluated with Analyst-Software. Percent bound is calculated
with the for-
mula: %bound = (plasma concentration ¨ buffer concentration/ plasma 30
concentration) X
100.
-46-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Evaluation of solubility
Saturated solutions are prepared in well plates (format depends on robot) by
adding an ap-
propriate volume of selected aqueous media (typically in the range of 0.25 -
1.5 ml) into
each well which contains a known quantity of solid drug substance (typically
in the range
.. 0.5 - 5.0 mg). The wells are shaken or stirred for a predefined time period
(typically in a
range of 2 - 24 h) and than filtered using appropriate filter membranes
(typically PTFE-fil-
ters with 0.45 p.m pore size). Filter absorption is avoided by discarding the
first few drops
of filtrate. The amount of dissolved drug substance is determined by UV
spectroscopy. In
addition the pH of the aqueous saturated solution is measured using a glass-
electrode pH
meter.
Evaluation of pharmacokinetic characteristics in rodents
The test compound is administered either intravenously to fed rats or orally
to fasted rats.
Blood samples are taken at several time points post application of the test
compound, anti-
is coagulated and centrifuged.
The concentration of analytes - the administered compound and/or metabolites -
are quanti-
fied in the plasma samples. PK parameters are calculated using non compartment
methods.
AUC and Cmax are normalized to a dose of 1 [tmol/kg.
zo .. Evaluation of Metabolism in human hepatocytes in vitro
The metabolic pathway of a test compound is investigated using primary human
hepatocytes
in suspension. After recovery from cryopreservation, human hepatocytes are
incubated in
Dulbecco's modified eagle medium containing 5% human serum and supplemented
with 3.5
[tg glucagon/500m1, 2.5mg insulin/500m1 and 3.75mg/500m1 hydrocortisone.
25 .. Following a 30 min preincubation in a cell culture incubator (37 C, 10%
CO2), test com-
pound solution is spiked into the hepatocyte suspension to obtain a final cell
density of
1.0*106 to 4.0*106 cells/ml (depending on the metabolic turnover rate of the
compound ob-
served with primary human hepatocytes), a final test compound concentration of
10 M, and
a final DMSO concentration of 0.05%.
30 The cells are incubated for six hours in a cell culture incubator on a
horizontal shaker, and
samples are removed from the incubation after 0, 0.5, 1, 2, 4 or 6 hours,
depending on the
metabolic turnover rate. Samples are quenched with acetonitrile and pelleted
by centrifuga-
tion. The supernatant is transferred to a 96-deepwell plate, evaporated under
nitrogen and
-47-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
resuspended prior to bioanalysis by liquid chromatography-high resolution mass
spectrom-
etry for identification of putative metabolites.
The structures are assigned tentatively based on Fourier-Transform-MS data.
Metabolites
are reported as percentage of the parent in human hepatocyte incubation with a
threshold of
4%.
-48-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
METHOD OF TREATMENT
The present invention is directed to compounds of general formula 1 which are
useful in
the prevention and/or treatment of a disease and/or condition associated with
or modulated
by TRPA1 activity, including but not limited to the treatment and/or
prevention of fibrotic
disease, inflammatory and immunoregulatory disorders, respiratory or
gastrointestinal dis-
eases or complaints, ophthalmic diseases, inflammatory diseases of the joints
and inflam-
matory diseases of the nasopharynx, eyes, and skin and pain and neurological
disorders.
Said disorders, diseases and complaints include cough, idiopathic pulmonary
fibrosis, other
io .. pulmonary interstitial diseases and other fibrotic, asthma or allergic
diseases, eosinophilic
diseases, chronic obstructive pulmonary disease, as well as inflammatory and
immunoreg-
ulatory disorders, such as rheumatoid arthritis and atherosclerosis, as well
as pain and neu-
rological disorders, such as acute pain, surgical pain, chronic pain and
depression and blad-
der disorders.
is The compounds of general formula 1 are useful for the prevention and/or
treatment of:
(1) Cough such as chronic idiopathic cough or chronic refractory cough, cough
associated
with asthma, COPD, lung cancer,post-viral infection and idiopathic pulmonary
fibrosis and
other pulmonary interstitial diseases.
(2) Pulmonary fibrotic diseases such as pneumonitis or interstitial
pneumonitis associated
zo with collagenosis, e.g. lupus erythematodes, systemic scleroderma,
rheumatoid arthritis,
polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as
pulmonary
lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory
bronchiolitis associated
interstitial lung disease, desquamative interstitial pneumonia, cryptogenic
orgainizing
pneumonia, acute interstitial pneumonia and lymphocytic interstitial
pneumonia, lymangi-
25 oleiomyomatosis, pulmonary alveolar proteinosis, Langerhan's cell
histiocytosis, pleural
parenchymal fibroelastosis, interstitial lung diseases of known cause, such as
interstitial
pneumonitis as a result of occupational exposures such as asbestosis,
silicosis, miners lung
(coal dust), farmers lung (hay and mould), Pidgeon fanciers lung (birds) or
other occupa-
tional airbourne triggers such as metal dust or mycobacteria, or as a result
of treatment
30 such as radiation, methotrexate, amiodarone, nitrofurantoin or
chemotherapeutics, or for
granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss
syndrome,
sarcoidosis, hypersensitivity pneumonitis, or interstitial pneumonitis caused
by different
-49-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis or
pneumonitis or inter-
stitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy,
M. boeck or
sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, or alpha-l-
antitrypsin defi-
ciency.
(3) Other fibrotic diseases such as hepatic bridging fibrosis, liver
cirrhosis, non-alcoholic
steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old
myocardial infarction,
glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture,
keloid, sclero-
derma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's
disease, nephro-
genic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.
II) (4) Inflammatory, auto-immune or allergic diseases and conditions such
as allergic or non-
allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal
polyposis, chronic rhinosi-
nusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis,
alveolitis, hyper-
reactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory
distress syn-
drome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic
cellulites
is (e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's
syndrome, chronic eosin-
ophilic pneumonia), eosinophilic fasciitis (e. g., Shulman's syndrome),
delayed-type hyper-
sensitivity, non-allergic asthma; exercise induced bronchoconstriction;
chronic obstructive
pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough,
pulmonary em-
physema; systemic anaphylaxis or hypersensitivity responses, drug allergies
(e.g., to peni-
20 cillin, cephalosporin), eosinophiliamyalgia syndrome due to the
ingestion of contaminated
tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid
arthritis,
Graves' disease, Sjogren's syndrome psoriatic arthritis, multiple sclerosis,
systemic lupus
erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, neonatal
throm-
bocytopenia, pediatric ITP), immune hemolytic anemia (auto-immune and drug
induced),
25 .. Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of
the newborn,
Goodpasture's syndrome (anti-GBM disease), Celiac, autoimmune cardio-myopathy
juve-
nile onset diabetes; glomerulonephritis, autoimmune thyroiditis, Behcet's
disease; graft re-
jection (e.g., in transplantation), including allograft rejection or
graftversus-host disease;
inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis;
spondyloar-
30 thropathies; scleroderma; psoriasis (including T-cell mediated
psoriasis) and inflammatory
dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact
dermatitis, ur-
ticaria; vasculitis (e. g., necrotizing, cutaneous, and hypersensitivity
vasculitis); erythema
nodosum; eosinophilic myositis, eosinophilic fasciitis, cancers with leukocyte
infiltration
-50-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
of the skin or organs; ophthalmic diseases such as age related macular
degeneration, dia-
betic retinopathy and diabetic macular edema, keratitis, eosinophilic
keratitis, keratocon-
junctivitis, vernal keratoconjunctivitis, scarring, anterior segment scarring,
blepharitis, ble-
pharoconjunctivitis, bullous disorders, cicatricial pemphigoid, conjunctival
melanoma, pa-
s pillary conjunctivitis, dry eye, episcleritis, glaucoma, gliosis,
Granuloma annulare, Graves'
ophthalmopathy, intraocular melanoma, Pinguecula, proliferative
vitreoretinopathy, pter-
ygia, scleritis, uveitis, acute gout flares, gout or osteoarthritis.
(5) Pain such as chronic idiopathic pain syndrome, neuropathic pain,
dysesthesia, allo-
dynia, migraine, dental pain and post-surgical pain.
II) (6) Depression, anxiousness, diabetic neuropathy and bladder disorders
such as bladder
outlet obstruction, overactive bladder, cystitis; myocardial reperfusion
injury or brain is-
chaemia injury.
Accordingly, the present invention relates to a compound of general formula 1
for use as a
medicament.
is .. Furthermore, the present invention relates to the use of a compound of
general formula 1
for the treatment and/or prevention of a disease and/or condition associated
with or modu-
lated by TRPA1 activity.
Furthermore, the present invention relates to the use of a compound of general
formula 1
for the treatment and/or prevention of fibrotic disease, inflammatory and
immunoregula-
20 tory disorders, respiratory or gastrointestinal diseases or complaints,
ophthalmic diseases,
inflammatory diseases of the joints and inflammatory diseases of the
nasopharynx, eyes,
and skin, pain and neurological disorders. Said disorders, diseases and
complaints include
cough, idiopathic pulmonary fibrosis, other pulmonary interstitial diseases
and other fi-
brotic, asthma or allergic diseases, eosinophilic diseases, chronic
obstructive pulmonary
25 disease, as well as inflammatory and immunoregulatory disorders, such as
rheumatoid ar-
thritis and atherosclerosis, as well as pain and neurological disorders, such
as acute pain,
surgical pain, chronic pain and depression and bladder disorders.
Furthermore, the present invention relates to the use of a compound of general
formula 1
for the treatment and/or prevention of:
30 (1) Cough such as chronic idiopathic cough or chronic refractory cough,
cough associated
with asthma, COPD, lung cancer,post-viral infection and idiopathic pulmonary
fibrosis and
other pulmonary interstitial diseases.
-51-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
(2) Pulmonary fibrotic diseases such as pneumonitis or interstitial
pneumonitis associated
with collagenosis, e.g. lupus erythematodes, systemic scleroderma, rheumatoid
arthritis,
polymyositis and dermatomysitis, idiopathic interstitial pneumonias, such as
pulmonary
lung fibrosis (IPF), non-specific interstitial pneumonia, respiratory
bronchiolitis associated
.. interstitial lung disease, desquamative interstitial pneumonia, cryptogenic
orgainizing
pneumonia, acute interstitial pneumonia and lymphocytic interstitial
pneumonia, lymangi-
oleiomyomatosis, pulmonary alveolar proteinosis, Langerhan's cell
histiocytosis, pleural
parenchymal fibroelastosis, interstitial lung diseases of known cause, such as
interstitial
pneumonitis as a result of occupational exposures such as asbestosis,
silicosis, miners lung
io (coal dust), farmers lung (hay and mould), Pidgeon fanciers lung (birds)
or other occupa-
tional airbourne triggers such as metal dust or mycobacteria, or as a result
of treatment
such as radiation, methotrexate, amiodarone, nitrofurantoin or
chemotherapeutics, or for
granulomatous disease, such as granulomatosis with polyangitis, Churg-Strauss
syndrome,
sarcoidosis, hypersensitivity pneumonitis, or interstitial pneumonitis caused
by different
is origins, e.g. aspiration, inhalation of toxic gases, vapors, bronchitis
or pneumonitis or inter-
stitial pneumonitis caused by heart failure, X-rays, radiation, chemotherapy,
M. boeck or
sarcoidosis, granulomatosis, cystic fibrosis or mucoviscidosis, or alpha-l-
antitrypsin defi-
ciency.
(3) Other fibrotic diseases such as hepatic bridging fibrosis, liver
cirrhosis, non-alcoholic
zo steatohepatitis (NASH), atrial fibrosis, endomyocardial fibrosis, old
myocardial infarction,
glial scar, arterial stiffness, arthrofibrosis, Dupuytren's contracture,
keloid, sclero-
derma/systemic sclerosis, mediastinal fibrosis, myelofibrosis, Peyronie's
disease, nephro-
genic systemic fibrosis, retroperitoneal fibrosis, adhesive capsulitis.
(4) Inflammatory, auto-immune or allergic diseases and conditions such as
allergic or non-
25 allergic rhinitis or sinusitis, chronic sinusitis or rhinitis, nasal
polyposis, chronic rhinosi-
nusitis, acute rhinosinusitis, asthma, pediatric asthma, allergic bronchitis,
alveolitis, hyper-
reactive airways, allergic conjunctivitis, bronchiectasis, adult respiratory
distress syn-
drome, bronchial and pulmonary edema, bronchitis or pneumonitis, eosinophilic
cellulites
(e.g., Well's syndrome), eosinophilic pneumonias (e.g., Loeffler's syndrome,
chronic eosin-
30 ophilic pneumonia), eosinophilic fasciitis (e. g., Shulman's syndrome),
delayed-type hyper-
sensitivity, non-allergic asthma; exercise induced bronchoconstriction;
chronic obstructive
pulmonary disease (COPD), acute bronchitis, chronic bronchitis, cough,
pulmonary em-
-52-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
physema; systemic anaphylaxis or hypersensitivity responses, drug allergies
(e.g., to peni-
cillin, cephalosporin), eosinophiliamyalgia syndrome due to the ingestion of
contaminated
tryptophane, insect sting allergies; autoimmune diseases, such as rheumatoid
arthritis,
Graves' disease, Sjogren's syndrome psoriatic arthritis, multiple sclerosis,
systemic lupus
erythematosus, myasthenia gravis, immune thrombocytopenia (adult ITP, neonatal
throm-
bocytopenia, pediatric ITP), immune hemolytic anemia (auto-immune and drug
induced),
Evans syndrome (platelet and red cell immune cytopaenias), Rh disease of the
newborn,
Goodpasture's syndrome (anti-GBM disease), Celiac, autoimmune cardio-myopathy
juve-
nile onset diabetes; glomerulonephritis, autoimmune thyroiditis, Behcet's
disease; graft re-
(e.g., in transplantation), including allograft rejection or graftversus-host
disease;
inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis;
spondyloar-
thropathies; scleroderma; psoriasis (including T-cell mediated psoriasis) and
inflammatory
dermatoses such as an dermatitis, eczema, atopic dermatitis, allergic contact
dermatitis, ur-
ticaria; vasculitis (e. g., necrotizing, cutaneous, and hypersensitivity
vasculitis); erythema
is nodosum; eosinophilic myositis, eosinophilic fasciitis, cancers with
leukocyte infiltration
of the skin or organs; ophthalmic diseases such as age related macular
degeneration, dia-
betic retinopathy and diabetic macular edema, keratitis, eosinophilic
keratitis, keratocon-
junctivitis, vernal keratoconjunctivitis, scarring, anterior segment scarring,
blepharitis, ble-
pharoconjunctivitis, bullous disorders, cicatricial pemphigoid, conjunctival
melanoma, pa-
pillary conjunctivitis, dry eye, episcleritis, glaucoma, gliosis, Granuloma
annulare, Graves'
ophthalmopathy, intraocular melanoma, Pinguecula, proliferative
vitreoretinopathy, pter-
ygia, scleritis, uveitis, acute gout flares, gout or osteoarthritis.
(5) Pain such as chronic idiopathic pain syndrome, neuropathic pain,
dysesthesia, allo-
dynia, migraine, dental pain and post-surgical pain.
(6) Depression, anxiousness, diabetic neuropathy and bladder disorders such as
bladder
outlet obstruction, overactive bladder, cystitis; myocardial reperfusion
injury or brain is-
chaemia injury.
In a further aspect the present invention relates to a compound of general
formula 1 for use
in the treatment and/or prevention of above mentioned diseases and conditions.
In a further aspect the present invention relates to the use of a compound of
general for-
mula 1 for the preparation of a medicament for the treatment and/or prevention
of above
mentioned diseases and conditions.
-53-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
In a further aspect of the present invention the present invention relates to
methods for the
treatment or prevention of above mentioned diseases and conditions, which
method com-
prises the administration of an effective amount of a compound of general
formula 1 to a
human being.
COMBINATION THERAPY
The compounds of the invention may further be combined with one or more,
preferably
one additional therapeutic agent. According to one embodiment the additional
therapeutic
to agent is selected from the group of therapeutic agents useful in the
treatment of diseases or
conditions described hereinbefore, in particular associated with fibrotic
diseases, inflam-
matory and immunoregulatory disorders, respiratory or gastrointestinal
diseases or com-
plaints, inflammatory diseases of the joints or of the nasopharynx, eyes, and
skin or condi-
tions such as for example cough, idiopathic pulmonary fibrosis, other
pulmonary intersti-
is tial diseases, asthma or allergic diseases, eosinophilic diseases,
chronic obstructive pulmo-
nary disease, atopic dermatitis as well as autoimmune pathologies, such as
rheumatoid ar-
thritis and atherosclerosis, or therapeutic agents useful for the treatment of
ophthalmic dis-
eases, pain and depression.
Additional therapeutic agents that are suitable for such combinations include
in particular
zo those, which, for example, potentiate the therapeutic effect of one or
more active sub-
stances with respect to one of the indications mentioned and/or allow the
dosage of one or
more active substances to be reduced.
Therefore, a compound of the invention may be combined with one or more
additional
therapeutic agents selected from the group consisting of antifibrotic agents,
anti-tussive
25 agents, anti-inflammatory agents, anti-atopic dermatitis agents,
analgesics, anti-convul-
sants, anxiolytics, sedatives, skeletal muscle relaxants or anti-depressants.
Antifibrotic agents are for example nintedanib, pirfenidone, phosphodiesterase-
IV (PDE4)
inhibitors such as roflumilast, autotaxin inhibitors such as GLPG-1690 or BBT-
877; con-
nective tissue growth factor (CTGF) blocking antibodies such as Pamrevlumab; B-
cell ac-
30 tivating factor receptor (BAFF-R) blocking antibodies such as Lanalumab;
alpha-V/beta-6
blocking inhibitors such as BG-00011/STX-100, recombinant pentraxin-2 (PTX-2)
such as
PRM-151; c-Jun N-terminal kinase (JNK) inhibitors such as CC-90001; galectin-3
inhibi-
tors such as TD-139; G-protein coupled receptor 84 (GPR84) inhibitors such as
GLPG-
-54-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
1205; G-protein coupled receptor 84/ G-protein coupled receptor 40 dual
inhibitors such as
PBI-4050; Rho Associated Coiled-Coil Containing Protein Kinase 2 (ROCK2)
inhibitors
such as KD-025; heat shock protein 47 (HSP47) small interfering RNA such as
BMS-
986263/ND-L02-s0201; Wnt pathway inhibitor such as SM-04646; LD4 / PDE3/4
inhibi-
s tors such as Tipelukast; recombinant immuno-modulatory domains of
histidyl tRNA syn-
thetase (HARS) such as ATYR-1923; prostaglandin synthase inhibitors such as ZL-
2102 /
SAR-191801; 15-hydroxy-eicosapentaenoic acid (15-HEPE e.g. DS-102); Lysyl
Oxidase
Like 2 (LOXL2) inhibitors such as PAT-1251, PXS-5382/PXS-5338;
phosphoinositide 3-
kinases (PI3K)/ mammalian target of rapamycin (mTOR) dual inhibitors such as
HEC-
io 68498; calpain inhibitors such as BLD-2660; mitogen-activated protein
kinase kinase ki-
nase (MAP3K19) inhibitors such as MG-S-2525; chitinase inhibitors such as OATD-
01;
mitogen-activated protein kinase-activated protein kinase 2 (MAPKAPK2)
inhibitors such
as MMI-0100; transforming growth factor beta 1 (TGF-betal) small interfering
RNA such
as TRK250/BNC-1021; or lysophosphatidic acid receptor antagonists such as BMS-
is 986278.
Anti-tussive agents are, for example, purinoceptor 3 (P2X3) receptor
antagonists such as
gefapixant, S-600918, BAY-1817080, or BLU-5937; neurokinin 1 (NK-1) receptor
antago-
nist such as Orvepitant, Aprepitant; nicotinic acetylcholine receptor alpha 7
subunit stimu-
lator such as ATA-101/bradanicline; codeine, gabapentin, pregablin, or
azithromycin.
zo Anti-inflammatory agents are, for example, corticosteroids such as
prednisolone or dexa-
methasone; cyclo-oxygenase-2 (COX2) inhibitors such as celecoxib, rofecoxib,
parecoxib,
valdecoxib, deracoxib, etoricoxib or lumiracoxib; prostaglandin E2
antagonists; leukotri-
ene B4 antagonists; leukotriene D4 antagonists such as monteleukast; 5-
lipoxygenase in-
hibitors; or other nonsteroidal anti-inflammatory agents (NSAIDs) such as
aspirin, diclo-
zs fenac, diflunisal, etodolac, ibuprofen or indomethacin.
Anti-atopic dermatitis agents are, for example, cyclosporin, methotrexate,
mycophenolate
mofetil, azathioprine, phosphodiesterase inhibitors (e.g. apremilast,
crisaborole), Janus As-
sociated Kinase (JAK) inhibitors (e.g. tofacitinib), neutralizing antibodies
against IL-4/IL-
13 (e.g. dupilamab), IL-13 (e.g. lebrikizumab, tralokinumab) and IL-31
(nemolizumab).
30 .. Analgesics are, for example, of the opioid type, such as morphine,
oxymorphine, levopa-
nol, oxycodon, propoxyphene, nalmefene, fentanyl, hydrocondon, hydromorphone,
meripidine, methadone, nalorphine, naloxone, naltrexone, buprenorphine,
butorphanol, nal-
buphine, pentazocine; or of the non-opioid type, such as acetophenamine.
-55-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Anti-depressants are, for example, tricyclic anti-depressants such as
amitriptyline, clomi-
pramine, despramine, doxepin, desipramine, imipramine, nortriptyline;
selective serotonin
reuptake inhibitor anti-depressants (SSRIs) such as fluoxetine, paroxetine,
sertraline, cital-
opram, escitalopram; norepinephrine reuptake inhibitor anti-depressants
(SNRIs) such as
maprotiline, lofepramine, mirtazapine, oxaprotiline, fezolamine, tomoxetine,
mianserin,
buproprion, hydroxybuproprion, nomifensine, viloxazine; dual serotonin-
norepinephrine
reuptake inhibitor anti-depressants (SNRIs) such as duloxetine, venlafaxine,
desvenlafax-
ine, levomilnacipran; atypical antidepressants such as trazodone, mirtazapine,
vortioxetine,
vilazodone, bupropion; or monoamine oxidase inhibitor anti-depressantss
(MAOIs) such as
tranylcypromine, phenelzine, or isocarboxazid.
Anxiolytics are, for example, benzodiazepines such as alprazolam, bromazepam,
chlordi-
azepoxide, clonazepam, clorazepate, diazepam, flurazepam, lorazepam, oxazepam,
temaze-
pam, triazolam, or tofisopam; or they are nonbenzodiazepine hypnoticssuch as
eszopi-
clone, zaleplon, zolpidem, or zopiclone; or they are carbamates e.g.
meprobamate, can-
is soprodol, tybamate, or lorbamate; or they are antihistamines such as
hydroxyzine, chlor-
pheniramine or diphenhydramine.
Sedatives are, for example, barbiturate sedatives, such as amobarbital,
aprobarbital, buta-
barbital, butabital, mephobarbital, metharbital, methohexital, pentobarbital,
secobarbital,
talbutal, theamylal, or thiopental; or they are non-barbiturate sedatives such
as glute-
n thimide, meprobamate, methaqualone or dichloalphenazone.
Skeletal muscle relaxants are, for example, baclofen, meprobamate,
carisoprodol, cyclo-
benzaprine, metaxalone, methocarbamol, tizanidine, chlorzoxazone or
orphenadrine.
Other suitable combination partners are inhibitors of Acetylcholinesterase
inhibitors such
as donepezil; 5-HT-3 anatgonists such as ondansetron; metabotropic glutamate
receptor an-
25 tagonists; antiarrhythmics such as mexiletine or phenytoin; or NMDA
receptor antagonists.
Further suitable combination partners are incontinence medications, for
example, anticho-
linergics such as oxybutynin, tolterodine, darifenacin, fesoterodine,
solifenacin or tro-
spium; or they are bladder muscle relaxants such as mirabegron; or they are
alpha blockers
such as tamsulosin, alfuzosin, silodosin, doxazosin or terazosin.
30 The dosage for the combination partners mentioned above is usually 1/5
of the lowest dose
normally recommended up to 1/1 of the normally recommended dose.
Therefore, in another aspect, this invention relates to the use of a compound
according to
the invention in combination with one or more additional therapeutic agents
described
-56-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
hereinbefore and hereinafter for the treatment of diseases or conditions which
may be af-
fected or which are mediated by TRPA1, in particular diseases or conditions as
described
hereinbefore and hereinafter.
In a further aspect this invention relates to a method for treating a disease
or condition
which can be influenced by the inhibition of TRPA1 in a patient that includes
the step of
administering to the patient in need of such treatment a therapeutically
effective amount of
a compound of formula (I) or a pharmaceutically acceptable salt thereof in
combination
with a therapeutically effective amount of one or more additional therapeutic
agents.
In a further aspect this invention relates to the use of a compound of formula
(I) or a phar-
maceutically acceptable salt thereof in combination with one or more
additional therapeu-
tic agents for the treatment of diseases or conditions which can be influenced
by the inhibi-
tion of TRPA1 in a patient in need thereof.
In yet another aspect the present invention relates to a method for the
treatment of a disease
or condition mediated by TRPA1 activity in a patient that includes the step of
administer-
is ing to the patient, preferably a human, in need of such treatment a
therapeutically effective
amount of a compound of the present invention in combination with a
therapeutically ef-
fective amount of one or more additional therapeutic agents described in
hereinbefore and
hereinafter.
The use of the compound according to the invention in combination with the
additional
zo therapeutic agent may take place simultaneously or at staggered times.
The compound according to the invention and the one or more additional
therapeutic
agents may both be present together in one formulation, for example a tablet
or capsule, or
separately in two identical or different formulations, for example as a so-
called kit-of-parts.
Consequently, in another aspect, this invention relates to a pharmaceutical
composition
25 that comprises a compound according to the invention and one or more
additional thera-
peutic agents described hereinbefore and hereinafter, optionally together with
one or more
inert carriers and/or diluents.
In yet another aspect the present invention relates to the use of a compound
according to
the invention in a cough-measuring device.
30 Other features and advantages of the present invention will become
apparent from the fol-
lowing more detailed examples which illustrate, by way of example, the
principles of the
invention.
-57-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
PREPARATION
The compounds according to the present invention and their intermediates may
be obtained
using methods of synthesis which are known to the one skilled in the art and
described in
the literature of organic synthesis. Preferably, the compounds are obtained in
analogous
fashion to the methods of preparation explained more fully hereinafter, in
particular as de-
scribed in the experimental section. In some cases, the order in carrying out
the reaction
steps may be varied. Variants of the reaction methods that are known to the
one skilled in
the art but not described in detail here may also be used.
The general processes for preparing the compounds according to the invention
will become
apparent to the one skilled in the art studying the following schemes. Any
functional
groups in the starting materials or intermediates may be protected using
conventional pro-
tecting groups. These protecting groups may be cleaved again at a suitable
stage within the
reaction sequence using methods familiar to the one skilled in the art.
is The compounds according to the invention are prepared by the methods of
synthesis de-
scribed hereinafter in which the sub stituents of the general formulae have
the meanings
given herein before. These methods are intended as an illustration of the
invention without
restricting its subject matter and the scope of the compounds claimed to these
examples.
Where the preparation of starting compounds is not described, they are
commercially ob-
tamable or may be prepared analogously to known compounds or methods described
herein. Substances described in the literature are prepared according to the
published meth-
ods of synthesis. Abbreviations are as defined in the Examples section.
Scheme 1:
-58-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0 K2CO3, 5 HCO2H 2Et3N
N, [cat.], ACN, RI
N
CI sN + LG.)(A DMA, RI Cf=-=-fN`NThrA _________
C1/ %.!KIMA
N¨NH N=14 0 N=N
HO
(A) (B)
0
0 0 RI %NANH2 0 0 Na0Et 0 0
Et0H R II NH3
Et0 OEt H .-
NH 1
y
neat, 100 C Et0 I OEt
80 C '`N Ni-i 100 C R1
II
OJ C)J
OEt RI
%N 0 Et0 0 H2N 0
H
(C) (D) (E)
N, 0
NANNNsN
N=14
HO (B)
. o.,t= NN'
HO
K2CO3, DMF
RI H2N 0 (I)
0 N
0 LION 0
RI%NANH CINA
MH:(B) RI%NAeN
',..õ..,N
N=N Me0H, R1, A .Nrs..N
THE, H20 N N -- sil
OtK2CO3, DMF (:)t N:"-N' .---- A ¨m- 0J\ N=1\11 .----
A
Et0 0 50 C to RI
HO HO
Et0 0 HO 'LO
(D) (F) (G)
0
NH2
R21 Ri 1\1)(N.N
_.
. NN'
DIPEA, HATU, o HO
DMF, RI
HNt 0
R21 (I)
In scheme 1, chloromethyltetrazole is N-alkylated with an appropriate ethanone
derivative
carrying a leaving group "LG" (e.g. Cl or Br) alpha to the carbonyl group in
the presence of
a base (e.g. K2CO3) to yield a mixture of two regioisomers. The undesired
regioisomer (not
shown) can be removed by chromatography using an appropriate gradient. The
resulting
ketone (A) can be reduced in an enantioselective fashion by using appropriate
catalytic sys-
tems using a transition metal complex (of e.g. Ru or Ir) in combination with a
chiral ligand
(e.g. ([(1S,2S)-2-amino-1,2-diphenylethyl](4-toluenesulfonyl)amido) and a
hydrogen source
such as formic acid triethylamine complex to yield alcohol (B).
io Uracil derivative (D) can be synthesized from a mono-substituted urea
and 1,3-diethyl 2-
(ethoxymethylidene)propanedioate under neat conditions at elevated temperature
to either
directly yield (D), or to yield (C), which can be further reacted to (D) under
basic conditions
such as Na0Et in Et0H at elevated temperature. Primary amide (E) can be
synthesized from
-59-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
ester (D) by stirring with ammonia in a solvent such as water or an alcohol at
elevated tem-
perature in a sealed vessel.
Final compounds (I) can be synthesized by alkylation of (E) with intermediate
(B) in pres-
ence of a base such as K2CO3. Alternatively, alkylation of intermediate (D)
with (B) in pres-
ence of a base gives (F), which can be hydrolysed with a suitable reagent such
as LiOH to
afford acid (G). Acid (G) can subsequently be coupled to an amine in the
presence of an
amide coupling reagent such as HATU and in the presence of a base such as
DIPEA to
provide final compounds (I).
Alternatively, compounds of formula (I) may be prepared as shown in Scheme 2
below.
Scheme 2:
0
0 0 NaN3NA
Ri %A LG Ri NEt NEt3 HCI
NH
1\1 NMP, 60 C
1\1 NH Nz.-N1
_______________________________________________________ 0
0 DIPEA 0
DMF, 88 C H2N 0
H2N 0 H2N 0
(E) (H)
0 0
LGNA 131,N AN
1\1)(Nr:-.%N.NH HO
0 0
DBU, NMP HO
H2N 0 RT H2N 0
(J) (I)
0
LG.)L 0
5 HCO2H 2Et3N
DIPEA 131.NAN.
THF, 70 C [cat.], THF, RT
0..)."====1=
0
H2N 0 (K)
In scheme 2, intermediate (H) can be prepared by alkylation of (E) with an
acetonitrile de-
rivative carrying a leaving group "LG" (e.g. Cl or Br) in presence of a base
such as DIPEA.
is Formation of the tetrazole (J) can be accomplished by typical reaction
conditions for te-
trazole formation (e.g. using NaN3 in the presence of TEA/TEA hydrochloride in
DMF).
-60-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Alkylation of the tetrazole (J) with an appropriate ethanone derivative
carrying a leaving
group "LG" (e.g. Cl or Br) alpha to the carbonyl group is run in presence of a
base such as
DIPEA to provide a mixture of two regioisomers. The undesired regioisomer (not
shown)
can be removed by chromatography using an appropriate gradient. Finally, the
keto group
of (K) can be reduced in an enantioselective fashion by using appropriate
catalytic systems
using a transition metal complex (of e.g. Ru or Ir) in combination with a
chiral ligand (e.g.
(R1S,2S)-2-amino-1,2-diphenylethylli4-toluenesulfonyl)amido) and a hydrogen
source such
as formic acid triethylamine complex to provide final compounds (I).
Alternatively, final
compounds (I) can be prepared by alkylation of intermediate (J) with an
appropriate aromatic
or heteroaromatic ethanol derivative carrying a leaving group "LG" (e.g. Cl or
Br) alpha to
the hydroxy group, in presence of a base such as DIPEA, and subsequent
isolation of the
desired regioisomer.
EXAMPLES
PREPARATION
The compounds according to the invention and their intermediates may be
obtained using
methods of synthesis which are known to the one skilled in the art and
described in the lit-
erature of organic synthesis for example using methods described in
"Comprehensive Or-
ganic Transformations", 2nd Edition, Richard C. Larock, John Wiley & Sons,
2010, and
"March's Advanced Organic Chemistry", 7th Edition, Michael B. Smith, John
Wiley &
Sons, 2013. Preferably the compounds are obtained analogously to the methods
of prepara-
tion explained more fully hereinafter, in particular as described in the
experimental section.
In some cases the sequence adopted in carrying out the reaction schemes may be
varied.
Variants of these reactions that are known to the skilled artisan but are not
described in de-
tail herein may also be used. The general processes for preparing the
compounds according
to the invention will become apparent to the skilled man on studying the
schemes that fol-
low. Starting compounds are commercially available or may be prepared by
methods that
are described in the literature or herein, or may be prepared in an analogous
or similar
manner. Before the reaction is carried out, any corresponding functional
groups in the
starting compounds may be protected using conventional protecting groups.
These protect-
ing groups may be cleaved again at a suitable stage within the reaction
sequence using
-61-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
methods familiar to the skilled man and described in the literature for
example in "Protect-
ing Groups", 3rd Edition, Philip J. Kocienski, Thieme, 2005, and "Protective
Groups in
Organic Synthesis", 4th Edition, Peter G. M. Wuts, Theodora W. Greene, John
Wiley &
Sons, 2006. The terms "ambient temperature" and "room temperature" are used
inter-
s changeably and designate a temperature of about 20 C, e.g. between 19
and 24 C.
Abbreviations:
ACN acetonitrile
Aq. aqueous
C Degree celsius
CyH/CH cyclohexane
conc. concentrated
DCM dichloro methane
DCE 1,2-Dichloroethane
DIPEA /V,N-diisopropylethylamine
DMA /V,N-dimethylacetamide
D 1VIF /V,N-dimethylformamide
DMSO dimethyl sulfoxide
ESI-MS Electrospray ionisation mass spectrometry
Et0Ac ethyl acetate
Et0H ethanol
ex example
eq equivalent
FA formic acid
hour
HATU 1-[Bis(dimethylamino)methylene]-1H-1,2,3-triazolo[4,5-
b]pyridinium
3-oxid hexafluorophosphate
HC1 Hydrochloric acid
HPLC High performance liquid chromatography
K2CO3 potassium carbonate
liter
molar
-62-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Me0H methanol
MgSO4 magnesium sulphate
min minute
mL milliliter
MTBE tert-butylmethylether
NH3 ammonia
RT room temperature (about 20 C)
sat. saturated
TBTU Benzotriazolyl tetramethyluronium tetrafluoroborate
TEA triethylamine
TFA trifluoroacetic acid
THF tetrahydrofuran
Preparation of Intermediates
Intermediate I
Intermediate 1.1 (general procedure)
2- [5- (chl oromethyl) -2H- 1,2,3,4 -tetrazol -2 -yl] -1- (4-
chlorophenyl)ethan - 1 - one
CI
CI
N\I CI
/
N-NH Br
0
0
To 1.00 g (8.44 mmol) 5-(chloromethyl)-2H-1,2,3,4-tetrazole and 2.17 g (9.28
mmol) 4-
chlorophenacyl bromide in 15 mL DMA are added 1.63 g (11.8 mmol) K2CO3 under
stirring
io at RT. The reaction mixture is stirred for 30 min at RT and subsequently
filtered. The filtrate
is diluted with water and sat. aq. NaCl-solution and is extracted with Et0Ac
three times. The
combined organic phases are washed with water, dried over Na2SO4, filtered
over activated
charcoal and the solvent is removed under reduced pressure. The residue is
purified by col-
umn chromatography (silica gel; CH/Et0Ac, 80/20 to 50/50 gradient) to provide
the product.
C10H8C12N40 (M = 271.1 g/mol)
ESI-MS: 271 [M+H]+
Rt (HPLC): 1.01 min (method B)
-63-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
The following compounds are prepared using procedures analogous to those
described for
intermediate 1.1 using appropriate starting materials. As is appreciated by
those skilled in the
art, these analogous examples may involve variations in general reaction
conditions.
1H NMR Reaction
(300 MHz, conditions
DMSO-d6) (deviation
Int. Starting materials Structure ESI-MS 6 ppm or from gen-
HPLC re- eral proce-
tention dure)
time [min]
(method)
5.10 (s,
2H), 6.59
(dd, J=
6.4, 0.8
Hz), 6.75
(m, 2H),
1.2 VIII.1 7.83 (d, J=
0
ci
8.03 (dd,
J= 8.7,
1.9Hz),
8.37 (d, J=
1.9 Hz,
1H)
ACN,
295 0.56 10min
1.3 VIII.2
[M+H]+ (A)
0
CI
Starting
materials
293 1.12 1:1
1.4
Br
[M+H]+ (B)
CI NN 0
2eq base
\ 311 1.23
1.5 0-1((1_13
\ I [m m+ (B)
0
CI
-64-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
I
I
\ \ 311/313 1.31
1 CI 01
1.6
c / [M+1-1]+ (B)
/
0 0
Stirred for
r lh;
r
315/317 1.00
CI
1.7
Br [M H]+ (H)
o
Starting
materials
1
1 311 1.08 1:1
1.8 a
Br \ [IVI H]F (H)
_-N 0
2 eq base,
starting
\L,)\]
Br 277 1.26
materials
1.9
o 0 [M-41]+ (B)
Stirred for
1:1
CI
15min
Starting
materials
F
295 1.02 1:1
1.10 VIII.4 cl
[M+H]+ (B)
CI
I
\ CI S \ 277 1.01
1.11
B [M+H]+ (H)
r /
I
0 0
0-----\
0
281 0.89
1.12 C\I Br [M+H]+ (H)
--A/ I
0 I\j\I 0
-65-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Starting
materials
295 1.02 1:1
1.13 VIII.3
ci [M+H]+ (B)
,ssf?1 251 0.95
1.14 Br [M+H]+ (H)
0 :-_--N 0
ACN,
//
stirred for
268 0.47 1.5h, puri-
1.15 VIII.5 S
[M+H]+ (G)
fled by
prep.
CI NN 0
HPLC
*see below
Br 267 0.75
table
1.16 11 0/ CI
0 [M+H]+ (C)
¨N 0
*p-methoxyphenacyl bromide (1.05 eq.) is slowly added to a stirred solution of
chlorome-
thyltetrazole and K2CO3 (1.4 eq) in DMA at 18 C; mixture is stirred at RT for
1.5 h; purifi-
cation via reversed phase HPLC (ACN/H20 gradient, 0.1% TFA).
Intermediate II
Intermediate 11.1 (general procedure)
(1R) - 2 - [5 - (chl oromethyl) - 2H- 1,2,3,4 -tetrazol -2 -yl] -1 - (4 - chl
orophenyl)ethan - 1 - ol
CI CI
N.
y y
CI N:--"N 0 CI N:--"N OH
1.30 g (4.80 mmol) 1-(4-chloropheny1)-2- [5-(chloromethyl)-2H-1,2,3,4-tetrazol
-2-
io yl]ethan-1-one (intermediate 1.1) is dissolved in 20 mL ACN under inert
atmosphere. 12
mg (0.02 mmol) Chloro([(1S,25)-2-amino-1,2-diphenylethyl](4-toluenesul-
fonyl)amido)(mesitylene)ruthenium (II) (CAS 174813-81-1) are added followed by
-66-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
dropwise addition of 0.72 mL (1.73 mmol) formic acid triethylamine complex
(5:2). After
stirring at RT for 3 h, the solvent is removed under reduced pressure. To the
remaining
crude mixture is added water and this mixture is extracted with Et0Ac. The
organic layers
are combined, dried over Na2SO4, filtered, treated with activated charcoal,
filtered and the
solvent is removed under reduced pressure to provide intermediate 11.1.
C10H10C12N40 (M = 273.1 g/mol)
ESI-MS: 273 [M+H]+
Rt (HPLC): 0.96 min (method B)
u) The following compounds are prepared using procedures analogous to those
described for
intermediate 11.1 using appropriate starting materials. As is appreciated by
those skilled in
the art, these analogous examples may involve variations in general reaction
conditions.
HPLC retention
Starting
Int. Structure ESI-MS time [min]
materials
(method)
N, 0 11.2 1.2 341 2.63
N
N=N HO F [M+HCO2]- (J)
11.3 1.3 297 0.52
(Ls-
CI HO [M+H]+ (A)
295 1.10
11.4 1.4 N,
N [M+H]+ (B)
CI NN OH
\
313 1.17
I1.5 1.5
[M+H]+ (B)
HO S
CI
-67-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
yi
CI 1--ci 313/315 1.26
11.6 1.6
/ S [M+H]+ (B)
I
OH
0 Br
317/319 1.14
11.7 1.7 N
-y [M+H]+ (B)
CI µN:_--N OH
0 CI
313 1.03
11.8 1.8 N.,..
/---- y [M+H]+ (B)
CI N---:N OH
Nr-
279 1.14
11.9 1.9 /
HO 0 [M+H]+ (B)
CI
O . F 297 0.97
11.10 1.10 N,
/--- y [M+H]+ (B)
CI N-----N OH
ytl-C1 279 0.97
H,
I 1 1.1 1 NI;
7-----e - s
CI \N-:-.-_N OH [M+H]+ (H)
0
0 0) 283 0.43
11.12 1.12 N
,....e-y [M+H]+ (A)
Cl \N-_:--N OH
F
0 297 0.97
11.13 1.13
N [M+H]+ (B)
Ci V-.--N OH
-68-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
11.14 1.14 253 0.48
[M+H]P (A)
CI N-.---N OH
270 0.41
11.15 1.15 S
N, OH [M+HIP
(A)
y
CI
11.16 1.16 269 0.45
1111\1 [M+H]P (G)
HO
Intermediate III
3-methy1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide
H N H N N H
0 0
N 0
/N
0 / 0
0
10.0 g (50.46 mmol) ethyl 3-methy1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-
carboxylate
(CAS: 154942-22-0, intermediate XII.6) in 33% aq. ammonia (120 mL) are stirred
in a
sealed vessel at 100 C for 10 h. The reaction mixture is cooled to RT and
concentrated under
reduced pressure. The residue is triturated with ACN, filtered off, and dried
at 50 C to pro-
vide intermediate III.
io C6H7N3 03 (M= 169.1 g/mol)
ESI-MS: 170 [M+H]P
Rt (HPLC): 0.48 min (method B)
Intermediate IV
is Intermediate IV.1 (general procedure)
-69-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
1- (5,6 - difluoro - 1 -b enzofuran - 2 - yl)ethan - 1 - one
0
HO 0 0
5.00 g (31.6 mmol) 4,5-difluoro-2-hydroxybenzaldehyde in 50 mL acetone is
treated with
6.99 g (50.6 mmol) potassium carbonate under argon at 0 C. After additional
stirring for 10
min at 0 C, 3.78 mL (47.4 mmol) chloroacetone are added dropwise and the
reaction mixture
is stirred at 70 C for 3 h. The reaction mixture is cooled to RT and
concentrated. The crude
is extracted with Et0Ac/water and the organic phase is concentrated under
reduced pressure
to provide intermediate IV.1.
Ci0H6F202 (M = 196.2 g/mol)
io 1H NMIR (300 MHz, DMSO-d6) 6 ppm: 2.56(s, 3 H), 7.89 (m, 1 H), 7.92 (m,
1 H), 8.01 (m,
1H)
The following compounds are prepared using procedures analogous to those
described for
intermediate IV.1 using appropriate starting materials. As is appreciated by
those skilled in
is the art, these analogous examples may involve variations in general
reaction conditions.
1H NMR Reaction
(300 MHz, conditions
DMSO-d6) 6 (deviation
Starting ma-
Int. Structure ESI-MS ppm or from general
teri al s HPLC reten- procedure)
tion time
[min]
(method)
Stirred at
179 0.50 90 C for
lh
IV.2 HO 41
0 0
[M+H]+ (A)
0-
0 1.1 eq chlo-
179 0.95 roacetone;
IV.3
0 0 F [M+H]- (B) 1.7 eq
K2CO3
-70-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
Solvent:
1.35 (t, J=
D1VIF ;
7.1 Hz, 3 H),
4
Br 10 0 T 1.0 eq bromo
.38 (q,'' acetic acid
Br 0 7.1 Hz, 2 H),
IW OH \ 7.68 (dd, J= ethyl ester
IV.4 ,o 0 0 -- 8.8, 2.1 Hz, 1 instead of
Br -/ chloroaceton
o H), 7.72-7.79
e; 1.5 eq.
1 (m, 2 H), N.
T,. 2k. c,c.J,
A3
8.04 (dd, J= stirred 'at
2.1, 0.6 Hz, 1
92 C over-
H)
night
o 1 1.1 eq chlo-
F
179 0.95
Is F / roacetone;
IV.5
0 0 [M+H]P (B) 1.7 eq
K2CO3
HO
Intermediate V
5-bromo-1-benzofuran-2-carboxylic acid
Br 0
\ Br 0
\
0 OH
To 6.58 g (24.4 mmol) ethyl 5-bromo-1-benzofuran-2-carboxylate (IV.4) in 3 mL
Et0H, 66
ml THF and 33 mL water are added 1.23 g (29.3 mmol) LiOH*H20 at 0 C. The
reaction
mixture is stirred at RT for 2 h and subsequently concentrated under reduced
pressure. The
residue is acidified with 1M HC1 to pH 5 and the resulting precipitate is
filtered off and dried
to provide intermediate V.
io C9H5BrO3 (M = 241.0 g/mol)
41 NMR (300 MHz, DM50-d6) 6 ppm: 7.59-7.76 (m, 3 H), 8.02 (d, J= 2.0 Hz, 1 H),
13.5 ¨
14.2 (br s, 1 H).
Intermediate VI
is 5-bromo-2-fluoro-1-benzofuran
Br \ 0 Br
_. \ F
0 OH
0
-71-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
5.00 g (20.7 mmol) 5-bromo-1-benzofuran-2-carboxylic acid (V), 14.70 g (41.5
mmol) Se-
lectflour and 4.82 g (83.0 mmol) potassium fluoride in 185 ml DCE and 95 ml
water are
stirred in a sealed tube at 70 C for 20 h. Subsequently, the reaction mixture
is extracted with
DCM/water. The organic layer is washed with brine, dried over Na2SO4 and
concentrated
under reduced pressure. The residue is purified by column chromatography
(silica gel,
DCM).
C8H4BrF0 (M = 215.0 g/mol)
NMR (300 MHz, DM50-d6) 6 ppm: 6.36 (dd, J= 6.4, 0.9 Hz, 1 H), 7.47 (dd, J=8.7,
2.1
Hz, 1 H), 7.58 (d, J= 8.7Hz, 1 H), 7.82 (d, J= 2.1 Hz, 1 H)
Intermediate VII
1-(2-fluoro-1-benzofuran-5-yl)ethan-1-one
0
Br
F F
0 0
To 218 mg (1.0 mmol) 5-bromo-2-fluoro-1-benzofuran (VI) in 3 mL DMF and 0.3 mL
water
is .. are added 168 mg (1.2 mmol) potassium carbonate under stirring at RT.
The mixture is
purged with argon followed by addition of 25 mg (0.1 mmol) 1,3-bis(diphe-
nylphosphino)propane, dppp, 7 mg palladium(II) acetate and 183 mg (2.5 mmol)
ethyl vinyl
ether. The reaction mixture is stirred at 80 C overnight, then cooled to RT
and treated with
aq. 1M HC1 (20mL). After stirring at RT for 30 min, the mixture is extracted
with Et0Ac
zo and the combined organic layers are concentrated under reduced pressure.
The crude product
is purified by column chromatography (silica gel; Et0Ac/hexane, gradient).
C 0H7F 02 (M = 178.2 g/mol)
NMR (300 MHz, DMSO-d6) 6 ppm: 2.63 (s, 3H), 6.49 (dd, J= 6.4, 0.8 Hz, 1H),
7.70
(dt, J= 8.7, 0.8 Hz, 1H), 7.93 (dd, J=8.7, 1.9 Hz, 1H), 8.25 (dd, J= 1.9, 0.6
Hz, 1H)
Intermediate VIII
Intermediate VIII.1 (general procedure)
-72-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
2-bromo-1-(2-fluoro-1-benzofuran-5-yl)ethan-1-one
0 0
B r
F F
0 0
126 mg (0.71 mmol) 1-(2-fluoro-1-benzofuran-5-yl)ethan-1-one (VII) in 1.5 mL
THF are
treated dropwise with 0.34 g (0.71 mmol) tetrabutylammonium tribromide in 0.08
mL
Me0H and 0.8 mL THF under stirring at RT. After stirring for 2 h, the reaction
mixture is
concentrated under reduced pressure and the residue is extracted with
Et0Ac/water. The
organic layer is concentrated under reduced pressure and the crude product is
purified by
column chromatography (silica gel; Hexane/Et0Ac, gradient).
CioH6BrF02 (M = 257.1 g/mol)
io 1I-INMR (300 MHz, DMSO-d6) 6 ppm: 4.99 (s, 2H), 6.53 (dd, J= 6.4, 0.9
Hz, 1H), 7.75 (d,
J= 8.7, 1H), 7.88-8.03 (m, 1H), 8.31 (dd, J= 1.9, 0.6 Hz, 1H)
The following compounds are prepared using procedures analogous to those
described for
intermediate VIII.1 using appropriate starting materials. As is appreciated by
those skilled
is .. in the art, these analogous examples may involve variations in general
reaction conditions.
1I-INMR (300 MHz, DMS0-
Starting ma- d) 6 ppm
Int. Structure ESI-MS or
terials
HPLC retention time [min]
(method)
257/259 0.58
VIII.2 0 o
IV.2 [M+H]+ (A)
Br
4.80 (s, 2H), 7.31 (ddd, J=
Br -\ 9.7, 8.8, 2.1 Hz, 1H),
7.73
VIII.3 IV.3 (dd, J= 9.1, 2.1 Hz, 1H),
7.93
0 (dd, J= 8.8, 5.7 Hz, 1H),
8.10
(d, J= 0.9 Hz, 1H)
Br -\ 257/259 1.03
VIII.4 IV.5
0 F [1\4+14]+ (B)
-73-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
VIII.5 s.õ'N 228/230 0.75
\ [M-H] (I)
0
*: The reaction is performed with bromine (13.6 eq) at RT for 2h in
dioxane/diethyl ether
and quenched with sodium thiosulfate solution.
Intermediate IX
.. ethyl 1-({2-[(2R)-2-(4-chloropheny1)-2-hydroxyethyl]-2H-1,2,3,4-tetrazol-5-
ylImethyl)-3-
methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate
CI
0 )LNO
N)cH
N=--N
HO
OH 0
0 0
CI 0
To 200 mg (1.01 mmol) ethyl 3-methy1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-
carbox-
ylate (CAS: 154942-22-0, intermediate XII.6) in 8 mL DMF are added 303 mg
(1.11
io mmol) of (1R)-2-[5 - (chloromethyl)-2H-1,2,3,4 -tetrazol -2 -yl] -1 - (4
- chloro-
phenyl)ethan-l-ol (intermediate 11.1) and 418 mg (3.03 mmol) K2CO3 and the
mixture is
stirred at 50 C for 5 h, then at RT for 17 h. The crude product is purified by
reversed phase
HPLC (ACN/H20 gradient, 0.1% TFA) to yield the desired product.
C18H19C1N605 (M = 434.8 g/mol)
is ESI-MS: 435 [M+H]+
Rt (HPLC): 0.48 min (method A)
Intermediate X
-74-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
1-({ 2- [(2R)-2-(4-chl oropheny1)-2-hy droxy ethyl] -2H-1,2,3 ,4-tetrazol-5-
ylIm ethyl)-3 -m e-
thy1-2,4-di oxo-1,2,3 ,4-tetrahy dropyrimi -carboxylicdine-5 acid
0 0
Th\I)LNr I Th\i)LN7ri\pl I
,...,. ¨1.-
HO
H
0 0
To 200 mg (0.46 mmol) intermediate IX in 1 mL methanol, 1 mL THF, and 100 1
water are
added 44 mg (1.8 mmol) lithium hydroxide. The reaction mixture is stirred for
1 h at 50 C
and subsequently cooled to RT and diluted with water. The aq. layer is washed
with DCM
three times, acidified with formic acid and the resulting precipitate is
filtered off and dried
at 50 C to yield the desired product.
C16H15C1N605 (M = 406.8 g/mol)
io ESI-MS: 407 [M+H]P
Rt (HPLC): 0.46 min (method A)
Intermediate XI
Intermediate XI.1 (general procedure)
is 1,3-diethyl 2-1 [(cyclobutylcarbamoyl)amino]methylidene}propanedioate
0
,
Th\IN H2 + 1
H 1 H
H
1.00 g (8.76 mmol) cyclobutylurea and 3.79 g (17.52 mmol) 1,3-diethyl 2-
(ethoxymethyli-
dene)propanedioate are heated under neat conditions at 100 C for 2.5 h, and at
130 C for 5
h. The reaction mixture is cooled to RT, diluted with methanol and purified by
reversed
zo phase HPLC (ACN/H20 gradient, 0.1% TFA) to provide intermediate XI.1.
C13H20N205 (M = 284.3 g/mol)
ESI-MS: 285 [M+H]P
Rt (HPLC): 0.53 min (method A)
-75-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
The following compounds are prepared using procedures analogous to those
described for
intermediate XI.1 using appropriate starting materials. As is appreciated by
those skilled in
the art, these analogous examples may involve variations in general reaction
conditions.
HPLC re- Reaction condi-
Int. Starting ma-
Structure ESI-MS tention
tions (deviation
terials
time [min] from general pro-
(method) cedure)
o o
2.5 h at 100 C, lh
259 0.47 at 130
C
XI.2 teN H2
[M+H]+ (A)
NK0
O o
1.5 h at 100 C
only
285 0.52
XI.3 XIV.1
[M+H]+ (A)
o o
3 h at 100 C only
287 0.58
XI.4 NI)LNH2
[M+H]+ (A)
O 0
3 h at 100 C only
XI.5 XIV.2 289 0.44
[M+H]+ (A)
Intermediate XII
Intermediate XII.1 (general procedure)
-76-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
ethyl 1-cyclobuty1-2-hydroxy-6-oxo-1,6-dihydropyrimidine-5-carboxylate
aNNH
0
0 0
To 2.00 g (7.03 mmol) intermediate XI.1 in 30 ml ethanol are added 957 mg
(14.1 mmol)
sodium ethoxide and the mixture is stirred at 80 C for 3 h, subsequently
diluted with ethanol
and purified by reversed phase HPLC (ACN/H20 gradient, 0.1% TFA).
C tHi4N204 (M = 238.2 g/mol)
ESI-MS: 239 [M+H]+
Rt (HPLC): 0.37 min (method A)
u) The following compounds are prepared using procedures analogous to those
described for
intermediate XII.1 using appropriate starting materials. As is appreciated by
those skilled in
the art, these analogous examples may involve variations in general reaction
conditions.
Reaction condi-
HPLC reten-
Int. Starting
Structure ESI-MS tion time [min] tions
(deviation
materials from general pro-
(method)
cedure)
HO 2 h at 80 C
XII.2 XI.2 213 0.28
[M+H]+ (A)
HO 4 h at 80 C;
addi-
XII.3 XI.3 239 0.36 tion
of 1 eq
Na-
[M+H]+ (A) OEt, 2 h at 80 C
HO 2 h at 80 C
241 0.40
XII.4 XI.4
[m+H]+ (A)
-77-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
2 h at 80 C
243 0.29
XII.5 XI.5
[M+H]P (A)
Intermediate XII.6
ethyl 3-methy1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxylate
0
0 0 0 NKN H
H2 1(3 (31
0 0
500 mg (6.75 mmol) methylurea and 1.36 g (6.75 mmol) 1,3-diethyl 2-
(methoxymethyli-
dene) propanedioate are stirred under neat conditions at 120 C for 2 h, at RT
for 17 h, at
100 C for 66 h, at 150 C for 17 h, and at 120 C for 17 h. Subsequently, the
mixture is diluted
with Et0Ac and refluxed. The mixture is slowly cooled to RT and the
precipitated interme-
diate is filtered off.
io C8H10N204 (M = 198.2 g/mol)
ESI-MS: 199 [M+H]P
Rt (HPLC): 0.24 min (method A)
Intermediate XIII
is Intermediate XIII.1 (general procedure)
1-cyclobuty1-2-hydroxy-6-oxo-1,6-dihydropyrimidine-5-carboxamide
Z
H2
0
0 0
630 mg (0.03 mmol) intermediate XII.1 in 10 ml aq. ammonia (33%) are stirred
at 85 C for
17 h in a sealed vessel. Stirring is continued at 100 C and aq. ammonia is
added until starting
-78-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
material has been consumed completely. Subsequently, the reaction mixture is
concentrated
under reduced pressure to provide intermediate XIII.1.
C9H1 iN303 (M = 209.2 g/mol)
ESI-MS: 210 [M+H]+
Rt (HPLC): 0.31 min (method A)
The following compounds are prepared using procedures analogous to those
described for
intermediate XIII.1 using appropriate starting materials. As is appreciated by
those skilled
in the art, these analogous examples may involve variations in general
reaction conditions.
HPLC retention
Starting materi-
Int. Structure ESI-MS time [min]
als
(method)
184 0.22
XIII.2 XII.2
H2
[M+H]+ (A)
0 0
XIII.3 210 0.32
XII.3 H2
[M+H]+ (A)
0 0
H
XIII.4 212 0.34
XII.4 1\1/\9\1I H2
** [M+H]+ (A)
0 0
HOc
XIII.5 214 0.22
XII.5 H2
** [M+H]+ (A)
0
0
io *workup: acidify with aq. HC1 (1M), extract with DCM, concentrate org
layer under re-
duced pressure, purification via reversed phase HPLC (ACN/H20 gradient, 0.1%
TFA).
** purification via reversed phase HPLC
Intermediate XIV.1
is (cyclopropylmethyl)urea
-79-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0
H C I
Fi2Nv
To 530 mg (4.93 mmol) 1-cyclopropylmethanamine hydrochloride in 2 ml water are
added
599 mg (7.39 mmol) potassium cyanate in small portions and the mixture is
stirred at 100 C
for 3 h. The reaction mixture is left at RT for 14 h, then intermediate XIV.1
is filtered off.
C5H10N20 (M = 114.2 g/mol)
ESI-MS: 115 [M+H]P
Rt (HPLC): 0.15 min (method A)
Intermediate XIV.2
io (2-methoxyethyl)urea
0
H2N H2NN
To 2.0 g (26.63 mmol) 2-methoxyethylamine in 8 ml water are added 3.24 g
(39.94 mmol)
potassium cyanate in small portions and the mixture is stirred at 100 C for 3
h, cooled to
RT, diluted with water/methanol and purified by reversed phase HPLC (ACN/H20
gradient,
is 0.1% TFA).
C4H10N202 (M = 114.2 g/mol)
ESI-MS: 119 [M+H]P
Rt (HPLC): 0.11 min (method A)
zo Preparation of Final Compounds
Example 1 (general procedure)
1412- [(2R)-2-(4-chl oropheny1)-2-hydroxy ethyl] -2H-1,2,3,4-tetrazol-5-
ylImethyl)-3 -me-
thy1-2,4-di oxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide
-80-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
CI
0
0
z N=N
HO
0 N- OH 0
0 H2 rc\I
0 H2
Ci
To 100 mg (0.59 mmol) 3-methy1-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-
carboxamide
(intermediate III) in 5 mL DMF are added 178 mg (0.65 mmol) (1R)-2-[5-
(chlorome-
thyl)-2H- 1,2,3,4 -tetrazol -2 -yl] - 1 - (4- chlorophenyl)ethan- 1-01
(intermediate 11.1) and
245 mg (1.77 mmol) K2CO3 and the mixture is stirred at RT overnight. The
mixture is pu-
rified by reversed phase HPLC (ACN/H20 gradient, 0.1% TFA) to yield the
desired prod-
uct.
Ci6Hi6C1N704 (M = 405.8 g/mol)
ESI-MS: 406 [M+H]+
io Rt (HPLC): 1.05 min (method B)
1H NMR (400 MHz, DMSO-d6) 6 ppm: 3.21 (s, 3 H), 4.73 -4.84 (m, 2 H), 5.13 (m,
1 H),
5.45 (s, 2 H), 5.92 (d, J=2.4 Hz, 1 H), 7.35-7.42 (m, 4 H), 7.64 (br d, J=3.4
Hz, 1 H), 8.20
(br d, J=3.4 Hz, 1 H), 8.77 (s, 1 H)
The following compounds are prepared using procedures analogous to those
described for
is example 1 using appropriate starting materials. As is appreciated by
those skilled in the art,
these analogous examples may involve variations in general reaction
conditions.
Starting Reaction
Ex. Structure
materials conditions
0
2 III + 11.4 2 eq. base
Solvent:
0
DMF, RT 22
HO S
0
H2
-81-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0
3 eq. base
Solvent:
3 III + 11.5 0 Ni¨ SF
DMF, RT
HO
overnight
0 H2
0 1.5
eq. base
Solvent:
4 III + 11.2
0 DMF,
RT
overnight
0 H2 HO
XN)%
Solvent:
III + 11.6 3 eq. base
i (II
DMF, RT
0 H2
HO S----Ncl
overnight
0
NAN 3 eq.
base
DSmolFve, nRtT
t :
0 I\L
6 III + 11.8 \IHO \
,
\
I
overnight
NH2
NN:(N
3 eq. base
7 III + 11.7 -----M____
Solvent:
0 DMF,
RT
r
HO
overnight
0 H2
-82-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0
NNAN 3 eq.
base
Solvent:
8 III + II. 1 0
Ot I\1 DMF, RT
HO
\
F
overnight
NNANO
3 eq. base
Solvent:
9 III + 11.9
0 4\1 DMA,
RT
\
HO
overnight
0 H2
NNAO N
3 eq. base
Solvent:
III + II. 1 1
Ni DMF, RT
HO S---Ncl
overnight
0 H2
1.5 eq. base
XN)0(N 0 Solvent:
11 III + 11.12 1
0 DMF, RT
HO
0 H2 overnight
0
\NA 1.5 eq. base
Solvent:
12 III + 11.13 DMF,
RT
Ot NH2 !-----)--->
NL
HO F overnight
0
0
3 eq. base
Solvent:
13 III + 11.14 0 N,
li DMF,
RT
0 H HO 2 overnight
-83-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0
1.5 eq. base
0XNAN--M______N .
Solvent:
14 III + 11.15
Ni¨ C DMF,
RT
HO S---
0 H2
overnight
NN
0
,NA 1.5
eq. base
Solvent:
15 III + 11.3 0 I\L /
overnight
DMF, RT
HO
0 N H2
F
0 2 eq.
base,
NNjci solvent:
16 III + 11.16 / DMF,
2 hat
0 *
50 C, RT
HO
H2
0
overnight
3 eq. base
Solvent: XIII.4 + )NNI)N
17 0 N, DMF,
RT 20
11.1 I
H 0 H2
h
NNN 3 eq. base
XIII.1 + Solvent:
18 0
11.1 * I DMF, RT 17
H 0 h H2
-84-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
0
3 eq. base
XIII.2 + /NNAN
Solvent:
19 0
II.1 N,
110 I
DMF, RT 17
HO
0 H2
3 eq. base
XIII.3 + Solvent:
0 N,
DMF, RT 17 11.1
0 H2
0
3 eq. base
XIII.5 + Solvent:
21 0
11.1 NN( DMF, RT 20
0 H2
Analytical data for the compounds described in the table above:
HPLC re-
tention time
Ex. ESI-MS 1H NMR (400 MHz, DM50-d6) 6 ppm
[min]
(method)
3.21 (s, 3 H), 4.87 - 5.03 (m, 2 H), 5.44 - 5.51 (m, 3
H), 6.44 (d, J=5.2 Hz, 1 H), 7.29 - 7.39 (m, 2 H),
428 0.45
2 7.32 (s, 1
H), 7.63 (d, J=3.5 Hz, 1 H), 7.74 - 7.78 (m,
[M+H]+ (A)
1 H), 7.93 (m, 1 H), 8.20 (d, J=3.5 Hz, 1 H), 8.79 (s,
1H)
3.21 (s, 3 H), 4.87 - 5.04 (m, 2 H), 5.42- 5.50 (m, 3
H), 6.46 (br d, J=4.7 Hz, 1 H), 7.22 (td, J=9.0, 2.5
446 1.07
3 Hz, 1 H),
7.31 (s, 1 H), 7.63 (d, J=3.5 Hz, 1 H), 7.78
[M+H]+ (B)
(dd, J8.7, 5.3 Hz, 1 H), 7.84 (dd, J9.3, 2.5 Hz, 1
H), 8.20 (d, J=3.5 Hz, 1 H), 8.77 (s, 1 H)
-85-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
3.22 (s, 3 H), 4.78 - 4.86 (m, 2 H), 5.20 (t, J=6.4 Hz,
1 H), 5.44 (s, 2 H), 5.7 - 6.1 (br s, 1 H), 6.30 (d,
430 0.44
4 J=6.3 Hz, 1 H), 7.30 (dd, J=8.6, 1.6 Hz, 1 H),
7.49
[M+H]P (A)
(d, J=8.6 Hz, 1 H), 7.59 (d, J=1.6 Hz, 1 H), 7.63 (d,
J=3.4 Hz, 1 H), 8.20 (d, J=3.4 Hz, 1 H), 8.76 (s, 1 H)
3.21 (s, 3 H), 4.80 - 5.00 (m, 2 H), 5.32 (m, 1 H),
446 1.13 5.46
(s, 2 H), 6.58 (d, J=5.3 Hz, 1 H), 7.10 (d, J=1.0
[M+H]P (B) Hz, 1
H), 7.62 (d, J=3.5 Hz, 1 H), 8.19 (d, J=3.5 Hz,
1 H), 8.79 (s, 1 H)
3.20 (s, 3 H), 4.97 - 5.10 (m, 2 H), 5.29 (m, 1 H),
5.44 (s, 2 H), 6.31 (s, 1 H), 6.83 (s, 1 H), 7.32 (dd,
446 1.09
6
J=8.7, 2.2 Hz, 1 H), 7.59 (d, J=8.7 Hz, 1 H), 7.63 (d,
[M+H]P (B)
J=3.4 Hz, 1 H), 7.68 (d, J=2.2 Hz, 1 H), 8.19 (d,
J=3.4 Hz, 1 H), 8.76 (s, 1 H)
3.22 (s, 3 H), 4.74 - 4.84 (m, 2 H), 5.11 (m, 1 H),
449/451 1.04 5.45
(s, 2 H), 5.92 (br s, 1 H), 7.33 (m, 2 H), 7.52 (m,
7
[M+H]P (B) 2 H), 7.63 (d, J=3.4 Hz, 1 H), 8.20 (d, J=3.4
Hz, 1
H), 8.77 (s, 1 H)
3.20 (s, 3 H), 4.97 - 5.10 (m, 2 H), 5.28(m, 1 H),
5.44 (s, 2 H), 6.29 (d, J=5.8 Hz, 1 H), 6.83 (s, 1 H),
430 1.09
8 7.12
(td, J=9.2, 2.7 Hz, 1 H), 7.41 (dd, J=8.9, 2.7 Hz,
[M+H]P (B)
1 H), 7.58 (dd, J=8.9, 4.2 Hz, 1 H), 7.62 (br d, J=3.2
Hz, 1 H), 8.19 (br d, J=3.2 Hz, 1 H), 8.75 (s, 1 H)
3.20 (s, 3 H), 4.98 - 5.09 (m, 2 H), 5.28 (m, 1 H),
5.44 (s, 2 H), 5.8 - 6.7 (br s, 1 H), 6.84 (s, 1 H), 7.23
412 0.53
9 [M+H] (D) (m, 1
H), 7.29 (m, 1 H), 7.55 (m, 1 H), 7.59 (m, 1 H),
P
7.62 (d, J=3.5 Hz, 1 H), 8.19 (d, J=3.5 Hz, 1 H), 8.77
(s, 1 H)
3.22 (s, 3 H), 4.80 - 4.92 (m, 2 H), 5.30 (m, 1 H),
412 1.03 5.45 (s, 2 H), 6.40 (br s, 1 H), 6.87 (dd, J= 3.8, 0.7
[M+H]P (B) Hz, 1 H), 6.96 (d, J=3.8 Hz, 1 H), 7.63 (br d,
J=3.3
Hz, 1 H), 8.19 (br d, J=3.3 Hz, 1H), 8.78(s, 1 H)
-86-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
3.22 (s, 3 H), 4.69 - 4.80 (m, 2 H), 4.99 - 5.07 (m, 1
H), 5.44 (s, 2 H), 5.76 (d, J=4.8 Hz, 1 H), 5.96 - 6.01
416 0.70
11 (m, 2H) 6.77 - 6.86 (m, 2H) 6.98(s 1H) 7.63 (br
[M+H]P (C)
d, J=3.1 Hz, 1 H), 8.20 (br d, J=3.1 Hz, 1 H), 8.78 (s,
1H)
3.20 (s, 3 H), 4.97 - 5.08 (m, 2 H), 5.26 (m, 1 H),
5.44 (s, 2 H), 6.26 (d, J=5.7 Hz, 1 H), 6.84 (s, 1 H),
430 0.77
12 7.12(m 1H) 7.51(m 1H) 7.60(m 1H) 7.62 (br
[M+H]P (C)
d, J=3.3 Hz, 1 H), 8.19 (br d, J=3.3 Hz, 1 H), 8.76 (s,
1H)
2.28 (s, 3 H), 3.22 (s, 3 H), 4.75 (m, 2 H), 5.07 (m, 1
13 386 1.02 H), 5.45 (s, 2 H), 5.75 (d, J=4.9 Hz, 1 H),
7.13 (m, 2
[M+H]P (B) H), 7.25 (m, 2 H), 7.64 (d, J=3.6 Hz, 1 H),
8.20 (d,
J=3.6 Hz, 1 H), 8.78 (s, 1 H)
3.22 (s, 3 H), 4.86 - 5.03 (m, 2 H), 5.45 (s, 2 H), 5.47
14 403 0.67 (m, 1 H), 6.70 (d, J=5.3 Hz, 1 H), 7.19 (d,
J=3.9 Hz,
[M+H]P (C) 1 H), 7.63 (br d, J=3.4 Hz, 1 H), 7.84 (d,
J=3.9 Hz, 1
H), 8.20 (br d, J=3.4 Hz, 1 H), 8.78 (s, 1 H)
3.20 (s, 3 H), 5.01 - 5.11 (m, 2 H), 5.32 (m, 1H),
15 430 0.55 5.44 (s, 2 H), 6.0 - 6.7 (br s, 1 H), 6.94 (m,
1 H), 7.17
[M+H]P (D) - 7.25 (m, 2 H), 7.43 (m, 1 H), 7.62 (d, J=3.5
Hz, 1
H), 8.19 (d, J=3.5 Hz, 1 H), 8.75 (s, 1 H)
3.22 (s, 3 H), 3.73 (s, 3H), 4.66 - 4.81 (m, 2 H), 5.05
16 402 0.37 (m, 1 H), 5.45 (s, 2 H), 5.71 (br s, 1 H), 6.88
(m, 2H),
[M+H]P (A) 7.29 (m, 2H), 7.64 (br d, J=3.1 Hz, 1 H), 8.20
(br d,
J=3.1 Hz, 1 H), 8.76 (s, 1 H)
0.83 (d, J=6.8 Hz, 6 H), 2.01 (m, 1 H), 3.70 (d, J=7.4
17 448 0.78 Hz, 2 H), 4.73 - 4.84 (m, 2 H), 5.11 (m, 1H),
5.44(s,
[M+H]P (I) 2 H), 5.5 - 6.3 (br s, 1 H), 7.38 (m, 4 H),
7.63 (br d,
J=3.4 Hz, 1 H), 8.20 (d, J=3.4 Hz, 1 H), 8.78 (s, 1 H)
1.62- 1.89(m, 2H), 2.11 - 2.24 (m, 2H), 2.68 -2.82
446 0.72
18 (m, 2H)' 4.74 - 4.85 (m, 2H)' 5.08-5.19(m' 2H)
[M+H]P (D)
5.40 (s, 2 H), 5.93 (d, J=4.8 Hz, 1 H), 7.38 (m, 4 H),
-87-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
7.60 (br d, J=3.4 Hz, 1 H), 8.16 (br d, J=3.4 Hz, 1
H), 8.73 (s, 1 H)
1.11 (t, J=7.0 Hz, 3 H), 3.88 (q, J=7.0 Hz, 2H), 4.73
19 420 0.67 - 4.86 (m, 2 H), 5.12(m, 1 H), 5.44 (s, 2 H), 5.5 -6.4
[M+H]P (E) (br s, 1 H), 7.34 - 7.44 (m, 4 H), 7.63 (br
d, J=3.3 Hz,
1 H), 8.20 (br d, J=3.3 Hz, 1 H), 8.76 (s, 1 H)
0.28 -0.46 (m, 4 H), 1.14 (m, 1 H), 3.75 (d, J=7.1
20 446 0.74 Hz, 2 H), 4.75 - 4.84 (m, 2 H), 5.12(m, 1 H), 5.45(s,
[M+H]P (I) 2 H), 5.92 (d, J=4.8 Hz, 1 H), 7.38 (m, 4 H),
7.64 (d,
J=3.4 Hz, 1 H), 8.19 (d, J=3.4 Hz, 1 H), 8.78 (s, 1 H)
3.22 (s, 3H), 3.49 (t, J=6.0 Hz, 2H), 4.04 (t, J=6.0
Hz, 2H), 4.74 -4.83 (m, 2 H), 5.12 (m, 1 H), 5.44 (s,
450 0.63
21 2 H), 5.6 - 6.2 (br s, 1 H), 7.34 - 7.42 (m, 4 H),
7.64
[M+H]P (I)
(br d, J=3.4 Hz, 1 H), 8.16 (br d, J=3.4 Hz, 1 H),
8.77 (s, 1 H)
Example 22 (general procedure)
1-(12-[(2R)-2-(4-chloropheny1)-2-hydroxyethyl]-2H-1,2,3,4-tetrazol-5-
ylImethyl)-N-(2-
methoxyethyl)-3-methyl-2,4-dioxo-1,2,3,4-tetrahydropyrimidine-5-carboxamide
CI CI
\o
N- N-
/
)\I
H2 T
= H NH
0 0
0
To 20 mg (0.05 mmol) intermediate X in 1.0 mL DMF are added 25 1 (0.15mmol)
DIPEA
and 22 mg (0.06 mmol) HATU under stirring at RT. After 30 min, 8p1(0.10 mmol)
2-meth-
oxyethan-l-amine are added and the mixture is stirred at RT for 90 min.
Subsequent purifi-
cation by reversed phase HPLC (ACN/H20 gradient, 0.1% TFA) yields the desired
product.
io C19H22C1N705 (M = 463.9 g/mol)
-88-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
ESI-MS: 464 [M+H]P
Rt (HPLC): 0.48 min (method A)
1E1 NMIR (400 MHz, DMSO-d6) 6 ppm 3.22 (s, 3 H), 3.28 (s, 3 H), 3.41 -3.51 (m,
4 H),
4.73 -4.84 (m, 2 H), 5.13 (m, 1 H), 5.45 (s, 2 H), 5.5 -6.4 (br s, 1 H), 7.38
(m, 4 H), 8.77
(s, 1 H), 8.94 (m, 1 H).
The following compounds are prepared using procedures analogous to those
described for
example 22 using appropriate starting materials. As is appreciated by those
skilled in the art,
these analogous examples may involve variations in general reaction
conditions.
Starting ma- Reaction
Ex. Structure
terials conditions
X + 0
23 H2
OH DMF, RT
90 min
F/\F
0
X +
0 /
=IN OH
DMF, RT
24
0
0
40 min
N H2
X 0
25 DMF, RT
)(:) 90 min
NH2 0
0
-89-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
I
X + 0 /
/
NH2 --N OH DMF, RT
1
26 ---j 60 min
I\ 0 F7111\1_)
0
I
x+ /
7----N OH DMF, RT
27
H2\ 40 min
0 /
H
0
0 CI
x+
5,N H2 1
=NI OH DMF, RT
28 7____)//0 90 min
0c) z.-,--0
0
H
0
I
x+
0 /
29
7----N OH 6eq base,
DMF, RT
0 60 min
1(1---\
H2N HCI 0
HCI
ON
-90-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
x+
0 CI
F-F---\ /
0 -------N OH DMF, RT
1-71---\ 90 mm
0 n
H2N 0
HCI (:).---
F
I
x+
31 F 0 /
F____ IN / DMF, RT
N=N OH
/ 90 min
H2N 0 Fc---F
0 F
I
X +
0 /
DMF, RT
32
.4
¨N OH
90 min
1-/
H2N 0 -1
0
x+ 0 CI
F 0 / 1\1
DMF, RT
33
/
---:---N OH
90 min
H2N
7---1
HCI 0
H
0
-91-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
X + 0
OH DMF,
RT
34
H)90 min
0
0
x+
OH DMF,
RT
overnight
0
H2N 0
Analytical data for the compounds described in the table above:
HPLC re-
Ex. ESI-MS tention time NMR (400 MHz, DMSO-d6) 6
ppm
[min]
(method)
3.23 (s, 3 H), 3.77 (m, 2 H), 4.74 - 4.84 (m, 2 H),
470 0.52 5.13 (m, 1 H), 5.46 (s, 2 H), 5.93 (br s, 1 H),
6.15 (tt,
23
[M+H]+ (A)
J=56.0, 3.7 Hz, 1 H), 7.38 (m, 4 H), 8.83 (s, 1 H),
9.07 (t, J=6.1 Hz, 1 H)
TFA salt: 3.12 (m, 2 H), 3.24 (s, 3 H), 3.32 (m, 2 H),
3.54 (m, 2 H), 3.65 (m, 2 H), 3.69 (m, 2 H), 3.99 (m,
519 0.38
24 2 H),
4.74 - 4.85 (m, 2 H), 5.13 (m, 1 H), 5.48 (s, 2
[M+H]+ (A)
H), 5.6 - 6.2 (br s, 1 H), 7.39 (m, 4 H), 8.82 (s, 1 H),
9.06 (t, J=6.1 Hz, 1 H), 9.54 (br s, 1 H)
TFA salt: 2.90 - 3.04 (br m, 3 H), 3.20 (m, 1 H), 3.25
519 0.38
25 (s, 3 H), 3.35-3.75 (br m, including solvent),
3.96 (m,
[M+H]+ (A)
1 H) 4.05 (m, 1 H), 4.74 - 4.85 (m, 2 H), 5.13 (m, 1
-92-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
H), 5.47 (s, 2 H), 5.9 (br s) 7.36-7.42 (m, 4 H) 8.83
(s, 1 H) 9.05 (t, J=6.1 Hz, 1 H) 9.68 (br s, 1 H)
3.23 (s, 3 H), 3.63 (s, 3 H), 4.55 (d, J=4.5 Hz, 2 H),
26 500 0.38 4.74 - 4.84 (m, 2 H), 5.13(m, 1 H), 5.46 (s,
2 H),
[M+H]P (A) 5.92 (d, J=4.9 Hz, 1 H), 6.82 (br s, 1 H), 7.10
(br s, 1
H), 7.38 (m, 4 H), 8.82 (s, 1 H), 9.34 (br t, 1 H)
2.82 (d, J=4.8 Hz, 3 H), 3.22 (s, 3 H), 4.74-4.84 (m,
27 420 0.46 2 H), 5.13 (m, 1 H), 5.45 (s, 2 H), 5.7 - 6.3
(br s, 1
[M+H]P (A) H), 7.38 (m, 4 H), 8.68 (q, J= 4.8 Hz, 1 H),
8.75 (s, 1
H)
3.03 (s, 3 H), 3.22 (s, 3 H), 3.37 (m, 2 H), 3.75 (m, 2
28 512 0.44 H), 4.73 -4.84 (m, 2 H), 5.13 (m, 1 H), 5.46
(s, 2 H),
[M+H]P (A) 5.93 (s, 1 H), 7.39 (m, 4 H), 8.80 (s, 1 H),
9.07 (t,
J=5.8 Hz, 1 H)
3.21 (s, 3 H), 3.66(m, 2 H), 4.15 (m, 2H), 4.74 -
4.84 (m, 2 H), 5.13 (m, 1 H), 5.45 (s, 2 H), 5.93 (d,
500 0.38
29 J=5.0 Hz, 1H) 6.88 (br s, 1H) 7.17 (br s, 1H)
7.38
[M+H]P (A)
(m, 4 H), 7.59 (br s, 1 H), 8.76 (s, 1 H), 8.86 (t, J=6.0
Hz, 1 H)
3.23 (s, 3 H), 3.64 (m, 2 H), 4.20 (t, J=5.4 Hz, 2 H),
30 518 0.84 4.75 -4.84 (m, 2 H), 5.13 (m, 1 H), 5.46 (s, 2
H), 5.5
[M+H]P (E) - 6.4 (br s, 1 H), 7.38 (m, 4 H), 8.80 (s, 1
H), 9.02 (t,
J=5.4 Hz, 1 H)
3.24 (s, 3 H), 4.19 (m, 2 H), 4.73-4.84 (m, 2H), 5.13
488 0.80
31 (m, 1H) 5.47(s 2H) 5.5 - 6.4 (br s, 1H) 7.38(m
[M+H]P (E)
4 H), 8.87 (s, 1 H), 9.22 (t, J=6.5 Hz, 1 H)
0.23 (m, 2 H), 0.45 (m, 2 H), 1.01 (m, 1 H), 3.18 (m,
32 460 0.80 2 H), 3.23 (s, 3 H), 4.73 - 4.84 (m, 2 H),
5.13 (m, 1
[M+H]P (E) H), 5.46 (s, 2 H), 5.6 - 6.2 (br s, 1 H), 7.38
(m, 4 H),
8.77 (s, 1 H), 8.90 (t, J=5.6 Hz, 1 H)
-93-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
3.23 (s, 3 H), 3.63 (dq, J=27.0, 5.1 Hz, 2 H), 4.53 (dt,
452 0.71 J=47.5, 5.1 Hz, 2 H), 4.73 - 4.84 (m, 2 H),
5.13(m, 1
33
[M+H]P (E) H), 5.46 (s, 2 H), 5.5 - 6.4 (br s, 1 H), 7.38
(m, 4 H),
8.80 (s, 1 H), 9.01 (t, J=5.8 Hz, 1 H)
1.11 (t, J=7.2 Hz, 3 H), 3.22 (s, 3 H), 3.32 (m, 2 H),
434 0.73 4.73 - 4.84 (m, 2 H), 5.13 (m, 1 H), 5.45 (s, 2
H), 5.5
34
[M+H]P (E) - 6.3 (br s, 1 H), 7.38 (m, 4 H), 8.75 (s, 1
H), 8.78 (t,
J=5.7 Hz, 1 H)
3.20 (s, 3 H), 3.72 (q, J=6.0 Hz, 2 H), 4.28 (t, J=6.0
Hz, 2 H), 4.74 - 4.84 (m, 2 H), 5.13 (m, 1 H), 5.45 (s,
500 0.87
35 2H) 6.24(t J=2.1 Hz, 1H) 7.38(m 4H) 7.47(m
[M+H]P (H)
1 H), 7.71 (m, 1 H), 8.77 (s, 1 H), 8.88 (t, J=6.0 Hz,
1H)
Analytical HPLC methods
Method A
Vol% water Flow
time (min) Vol% ACN
(incl. 0.1% TFA)
[mL/min]
0.00 99 1 1.6
0.02 99 1 1.6
1.00 0 100 1.6
1.10 0 100 1.6
Analytical column: )(Bridge BEH (Waters) C18 2.1 x 30 mm 1.711m; column
tempera-
ture: 60 C
Method B
Vol% water Flow
time (min) Vol% ACN
(incl. 0.1% TFA)
[mL/min]
0.00 97 3 2.2
0.20 97 3 2.2
1.20 0 100 2.2
-94-

CA 03181350 2022-10-26
WO 2022/002782 PCT/EP2021/067470
1.25 0 100 3.0
1.40 0 100 3.0
Analytical column: Stable Bond (Agilent) C18 3.0 x 30 mm 1.8[1..m; column
temperature:
60 C
Method C
Vol% water Flow
time (min) Vol% ACN
(incl. 0.1% NH3)
[mL/min]
0.00 97 3 2.2
0.20 97 3 2.2
1.20 0 100 2.2
1.25 0 100 3.0
1.40 0 100 3.0
Analytical column: Xbridge (Waters) C18 3.0 x 30 mm 2.5[tm; column
temperature:
60 C
Method D
Vol.% water
time (min) Vol. % ACN Flow [mL/min]
(incl. 0.1 % NH4OH)
0.00 95 5 1.5
1.30 0 100 1.5
1.50 0 100 1.5
1.60 95 5 1.5
Analytical column: )(Bridge C18 3.0 x 30 mm 2.5 p.m (Waters); column
temperature:
60 C
Method E
Vol% water Vol% ACN Flow
time (min)
(incl. 0.1% TFA) 0.08%TFA
[mL/min]
0.00 95 5 1.5
1.30 0 100 1.5
-95-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
1.50 0 100 1.5
1.60 95 5 1.5
Analytical column: Sunfire (Waters); C18 3.0 x 30 mm 2.5 p.m; column
temperature:
60 C
Method F
Vol% water Flow
time (min) Vol% ACN
(incl. 0.1% TFA)
[mL/min]
0.00 95 5 1.3
0.02 95 5 1.3
1.00 0 100 1.3
1.30 0 100 1.3
Analytical column: )(Bridge BEH (Waters) C18 2.1 x 30 mm 2.5pm; column tempera-
ture: 60 C
Method G
Vol.% water
time (min) Vol. % ACN Flow [mL/min]
(incl. 0.1 % TFA)
0.00 99 1 1.6
0.02 99 1 1.6
1.0 0 100 1.6
1.1 0 100 1.6
Analytical column: Zorbax StableBond C18 (Agilent) 1.8 m; 2.1 x 30 mm; column
tem-
perature: 60 C
Method H
Vol% water Flow
time (min) Vol% ACN
(incl. 0.1% TFA)
[mL/min]
0.00 97 3 2.2
0.20 97 3 2.2
-96-

CA 03181350 2022-10-26
WO 2022/002782
PCT/EP2021/067470
1.20 0 100 2.2
1.25 0 100 3.0
1.40 0 100 3.0
Analytical column: Sunfire (Waters) 2.5 p.m; 3.0 x 30 mm; column temperature:
60 C
Method I
Vol.% water
time (min) Vol. % ACN Flow [mL/min]
(incl. 0.1 % TFA)
0.00 95 5 1.5
1.30 0 100 1.5
1.50 0 100 1.5
Analytical column: Sunfire C18 (Waters) 2.5 p.m; 3.0 x 30 mm; column
temperature:
60 C
Method J
Vol.% water Vol% ACN Flow
time (min)
(incl. 0.1 % FA) (incl. 0,1% FA)
[mL/min]
0.00 60 40 0.5
6.00 40 60 0.5
6.8 40 60 0.5
7.00 10 90 0.5
8.10 10 90 0.5
8.50 60 40 0.5
60 40 0.5
Analytical column: Acquity UPLC BEH ; C8 2.1 x 150 mm 1.71.1.m; column
temperature:
55 C
5
-97-

Representative Drawing

Sorry, the representative drawing for patent document number 3181350 was not found.

Administrative Status

2024-08-01:As part of the Next Generation Patents (NGP) transition, the Canadian Patents Database (CPD) now contains a more detailed Event History, which replicates the Event Log of our new back-office solution.

Please note that "Inactive:" events refers to events no longer in use in our new back-office solution.

For a clearer understanding of the status of the application/patent presented on this page, the site Disclaimer , as well as the definitions for Patent , Event History , Maintenance Fee  and Payment History  should be consulted.

Event History

Description Date
Inactive: IPC assigned 2023-01-16
Inactive: IPC assigned 2023-01-16
Inactive: First IPC assigned 2023-01-09
Letter sent 2022-12-07
Compliance Requirements Determined Met 2022-12-05
Priority Claim Requirements Determined Compliant 2022-12-05
Application Received - PCT 2022-12-05
Inactive: IPC assigned 2022-12-05
Request for Priority Received 2022-12-05
National Entry Requirements Determined Compliant 2022-10-26
Application Published (Open to Public Inspection) 2022-01-06

Abandonment History

There is no abandonment history.

Maintenance Fee

The last payment was received on 2023-12-20

Note : If the full payment has not been received on or before the date indicated, a further fee may be required which may be one of the following

  • the reinstatement fee;
  • the late payment fee; or
  • additional fee to reverse deemed expiry.

Patent fees are adjusted on the 1st of January every year. The amounts above are the current amounts if received by December 31 of the current year.
Please refer to the CIPO Patent Fees web page to see all current fee amounts.

Fee History

Fee Type Anniversary Year Due Date Paid Date
Basic national fee - standard 2022-10-26 2022-10-26
MF (application, 2nd anniv.) - standard 02 2023-06-27 2022-10-26
MF (application, 3rd anniv.) - standard 03 2024-06-25 2023-12-20
Owners on Record

Note: Records showing the ownership history in alphabetical order.

Current Owners on Record
BOEHRINGER INGELHEIM INTERNATIONAL GMBH
Past Owners on Record
FLORIAN PAUL CHRISTIAN BINDER
JENS WILLWACHER
MARTIN THOMAS FLECK
Past Owners that do not appear in the "Owners on Record" listing will appear in other documentation within the application.
Documents

To view selected files, please enter reCAPTCHA code :



To view images, click a link in the Document Description column. To download the documents, select one or more checkboxes in the first column and then click the "Download Selected in PDF format (Zip Archive)" or the "Download Selected as Single PDF" button.

List of published and non-published patent-specific documents on the CPD .

If you have any difficulty accessing content, you can call the Client Service Centre at 1-866-997-1936 or send them an e-mail at CIPO Client Service Centre.


Document
Description 
Date
(yyyy-mm-dd) 
Number of pages   Size of Image (KB) 
Description 2022-10-25 97 2,787
Claims 2022-10-25 22 272
Abstract 2022-10-25 1 56
Courtesy - Letter Acknowledging PCT National Phase Entry 2022-12-06 1 595
Patent cooperation treaty (PCT) 2022-10-25 1 84
Declaration 2022-10-25 2 82
National entry request 2022-10-25 5 174
International search report 2022-10-25 2 59